1
|
Sinha S, Wal P, Goudanavar P, Divya S, Kimothi V, Jyothi D, Sharma MC, Wal A. Research on Alzheimer's Disease (AD) Involving the Use of In vivo and In vitro Models and Mechanisms. Cent Nerv Syst Agents Med Chem 2025; 25:123-142. [PMID: 38803173 DOI: 10.2174/0118715249293642240522054929] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Revised: 03/11/2024] [Accepted: 03/26/2024] [Indexed: 05/29/2024]
Abstract
BACKGROUND Alzheimer's Disease (AD) is a neurodegenerative disorder characterized by the progressive formation of extracellular amyloid plaques, intracellular neurofibrillary tangles, inflammation, and impaired antioxidant systems. Early detection and intervention are vital for managing AD effectively. OBJECTIVES This review scrutinizes both in-vivo and in-vitro screening models employed in Alzheimer's disease research. in-vivo models, including transgenic mice expressing AD-related mutations, offer profound insights into disease progression and potential therapeutic targets. A thorough understanding of these models and mechanisms will facilitate the development of novel therapies and interventions for Alzheimer's disease. This review aims to provide an overview of the current experimental models in AD research, assess their strengths and weaknesses as model systems, and underscore the future prospects of experimental AD modeling. METHODS We conducted a systematic literature search across multiple databases, such as Pub- Med, Bentham Science, Elsevier, Springer Nature, Wiley, and Research Gate. The search strategy incorporated pertinent keywords related to Alzheimer's disease, in-vivo models, in-vitro models, and screening mechanisms. Inclusion criteria were established to identify studies focused on in-vivo and in-vitro screening models and their mechanisms in Alzheimer's disease research. Studies not meeting the predefined criteria were excluded from the review. RESULTS A well-structured experimental animal model can yield significant insights into the neurobiology of AD, enhancing our comprehension of its pathogenesis and the potential for cutting-edge therapeutic strategies. Given the limited efficacy of current AD medications, there is a pressing need for the development of experimental models that can mimic the disease, particularly in pre-symptomatic stages, to investigate prevention and treatment approaches. To address this requirement, numerous experimental models replicating human AD pathology have been established, serving as invaluable tools for assessing potential treatments. CONCLUSION In summary, this comprehensive review underscores the pivotal role of in-vivo and in-vitro screening models in advancing our understanding of Alzheimer's disease. These models offer invaluable insights into disease progression, pathological mechanisms, and potential therapeutic targets. By conducting a rigorous investigation and evaluation of these models and mechanisms, effective screening and treatment methods for Alzheimer's disease can be devised. The review also outlines future research directions and areas for enhancing AD screening models.
Collapse
Affiliation(s)
- Sweta Sinha
- LCIT School of Pharmacy, Bilaspur, Chattisgarh, India
| | - Pranay Wal
- PSIT-Pranveer Singh Institute of Technology (Pharmacy) NH19 Kanpur Agra Highway, Bhaunti Kanpur, India
| | - Prakash Goudanavar
- Sri Adichunchanagiri College of Pharmacy, Adichunchanagiri University B.G.Nagara, Karnataka, India
| | | | | | - Divya Jyothi
- NGSM Institute of Pharmaceutical Sciences, Nitte University, Paneer Deralakatte, Mangaluru, 575018, India
| | | | - Ankita Wal
- PSIT-Pranveer Singh Institute of Technology (Pharmacy) NH19 Kanpur Agra Highway, Bhaunti Kanpur, India
| |
Collapse
|
2
|
Kshirsagar S, Islam MA, Reddy AP, Reddy PH. Cell culture research in aging and Alzheimer's disease: The strategic use/reuse of untreated controls and savings people's tax dollars. J Alzheimers Dis Rep 2025; 9:25424823241310716. [PMID: 40034533 PMCID: PMC11864248 DOI: 10.1177/25424823241310716] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Accepted: 12/04/2024] [Indexed: 03/05/2025] Open
Abstract
Cell culture is an essential tool in both fundamental and translational research, particularly for understanding complex diseases like Alzheimer's disease (AD). The use of cell lines provides the advantage of genetic homogeneity, ensuring reproducible and consistent results. This article explores the application of mammalian cell cultures to model AD, focusing on the transfection of cells with key genes associated with the disease to replicate the cellular environment of AD. It explains various transfection methods and challenges related to the process. These models offer a robust platform for investigating cellular biology, molecular pathways, physiological processes, and drug discovery efforts. A range of assays, including RT-PCR, western blotting, ELISA, mitochondrial respiration, and reactive oxygen species analysis, are employed to assess the impact of genetic modifications on cellular functions and to screen potential AD therapies. Researchers often design experiments with multiple variables such as genetic modifications, chemical treatments, or time points, paired with positive and negative controls. By using a consistent control group across all conditions and under identical experimental conditions, researchers can minimize variability and enhance data reproducibility. This approach is particularly valuable in AD research, where small experimental differences can significantly influence outcomes. Using a shared control group ensures data comparability across experiments, saving time and resources by eliminating redundant control tests. This strategy not only streamlines the research process but also improves the reliability of results, making it a sensible, resource-efficient method that ultimately conserves public funding in the pursuit of AD treatments.
Collapse
Affiliation(s)
- Sudhir Kshirsagar
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, USA
| | - Md Ariful Islam
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, USA
| | - Arubala P Reddy
- Nutritional Sciences Department, College of Human Sciences, Texas Tech University, Lubbock, TX, USA
| | - P Hemachandra Reddy
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, USA
- Nutritional Sciences Department, College of Human Sciences, Texas Tech University, Lubbock, TX, USA
- Department of Pharmacology and Neuroscience, Texas Tech University Health Sciences Center, Lubbock, TX, USA
- Department of Neurology, Texas Tech University Health Sciences Center, Lubbock, TX, USA
- Department of Public Health, Graduate School of Biomedical Sciences, Texas Tech University Health Sciences Center, Lubbock, TX, USA
- Department of Speech, Language, and Hearing Sciences, Texas Tech University Health Sciences Center, Lubbock, TX, USA
| |
Collapse
|
3
|
Sharlow ER, Llaneza DC, Grever WE, Mingledorff GA, Mendelson AJ, Bloom GS, Lazo JS. High content screening miniaturization and single cell imaging of mature human feeder layer-free iPSC-derived neurons. SLAS DISCOVERY : ADVANCING LIFE SCIENCES R & D 2023; 28:275-283. [PMID: 36273809 PMCID: PMC10119332 DOI: 10.1016/j.slasd.2022.10.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 09/27/2022] [Accepted: 10/11/2022] [Indexed: 11/05/2022]
Abstract
Human induced pluripotent stem cell (iPSC)-derived neurons are being increasingly used for high content imaging and screening. However, iPSC-derived neuronal differentiation and maturation is time-intensive, often requiring >8 weeks. Unfortunately, the differentiating and maturing iPSC-derived neuronal cultures also tend to migrate and coalesce into ganglion-like clusters making single-cell analysis challenging, especially in miniaturized formats. Using our defined extracellular matrix and low oxygen culturing conditions for the differentiation and maturation of human cortical neurons, we further modified neuronal progenitor cell seeding densities and feeder layer-free culturing conditions in miniaturized formats (i.e., 96 well) to decrease neuronal clustering, enhance single-cell identification and reduce edge effects usually observed after extended neuronal cell culture. Subsequent algorithm development refined capabilities to distinguish and identify single mature neurons, as identified by NeuN expression, from large cellular aggregates, which were excluded from image analysis. Incorporation of astrocyte conditioned medium during differentiation and maturation periods significantly increased the percentage (i.e., ∼10% to ∼30%) of mature neurons (i.e., NeuN+) detected at 4-weeks post-differentiation. Pilot, proof of concept studies using this optimized assay system yielded negligible edge effects and robust Z-factors in population-based as well as image-based neurotoxicity assay formats. Moreover, moxidectin, an FDA-approved drug with documented neurotoxic adverse effects, was identified as a hit using both screening formats. This miniaturized, feeder layer-free format and image analysis algorithm provides a foundational imaging and screening platform, which enables quantitative single-cell analysis of differentiated human neurons.
Collapse
Affiliation(s)
- Elizabeth R Sharlow
- Department of Pharmacology, University of Virginia, 340 Jefferson Park Avenue, Pinn Hall, 5th Floor, P.O. Box 800735, Charlottesville, VA 22908-0735, USA.
| | - Danielle C Llaneza
- Department of Pharmacology, University of Virginia, 340 Jefferson Park Avenue, Pinn Hall, 5th Floor, P.O. Box 800735, Charlottesville, VA 22908-0735, USA
| | | | - Garnett A Mingledorff
- Department of Pharmacology, University of Virginia, 340 Jefferson Park Avenue, Pinn Hall, 5th Floor, P.O. Box 800735, Charlottesville, VA 22908-0735, USA
| | - Anna J Mendelson
- Department of Pharmacology, University of Virginia, 340 Jefferson Park Avenue, Pinn Hall, 5th Floor, P.O. Box 800735, Charlottesville, VA 22908-0735, USA
| | - George S Bloom
- Department of Biology, University of Virginia, 420 Gilmer Hall, 485 McCormick Road, P.O. Box 400328, Charlottesville VA 22904, USA; Department of Cell Biology, University of Virginia, 420 Gilmer Hall, 485 McCormick Road, P.O. Box 400328, Charlottesville VA 22904, USA; Department of Neuroscience, University of Virginia, 420 Gilmer Hall, 485 McCormick Road, P.O. Box 400328, Charlottesville VA 22904, USA
| | - John S Lazo
- Department of Pharmacology, University of Virginia, 340 Jefferson Park Avenue, Pinn Hall, 5th Floor, P.O. Box 800735, Charlottesville, VA 22908-0735, USA
| |
Collapse
|
4
|
Unzueta-Larrinaga P, Barrena-Barbadillo R, Ibarra-Lecue I, Horrillo I, Villate A, Recio M, Meana JJ, Diez-Alarcia R, Mentxaka O, Segarra R, Etxebarria N, Callado LF, Urigüen L. Isolation and Differentiation of Neurons and Glial Cells from Olfactory Epithelium in Living Subjects. Mol Neurobiol 2023; 60:4472-4487. [PMID: 37118325 PMCID: PMC10293402 DOI: 10.1007/s12035-023-03363-2] [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: 01/17/2023] [Accepted: 04/19/2023] [Indexed: 04/30/2023]
Abstract
The study of psychiatric and neurological diseases requires the substrate in which the disorders occur, that is, the nervous tissue. Currently, several types of human bio-specimens are being used for research, including postmortem brains, cerebrospinal fluid, induced pluripotent stem (iPS) cells, and induced neuronal (iN) cells. However, these samples are far from providing a useful predictive, diagnostic, or prognostic biomarker. The olfactory epithelium is a region close to the brain that has received increased interest as a research tool for the study of brain mechanisms in complex neuropsychiatric and neurological diseases. The olfactory sensory neurons are replaced by neurogenesis throughout adult life from stem cells on the basement membrane. These stem cells are multipotent and can be propagated in neurospheres, proliferated in vitro and differentiated into multiple cell types including neurons and glia. For all these reasons, olfactory epithelium provides a unique resource for investigating neuronal molecular markers of neuropsychiatric and neurological diseases. Here, we describe the isolation and culture of human differentiated neurons and glial cells from olfactory epithelium of living subjects by an easy and non-invasive exfoliation method that may serve as a useful tool for the research in brain diseases.
Collapse
Affiliation(s)
- Paula Unzueta-Larrinaga
- Department of Pharmacology, University of the Basque Country, UPV/EHU, Leioa, Spain
- Biocruces Bizkaia Health Research Institute, Barakaldo, Spain
| | - Rocío Barrena-Barbadillo
- Biocruces Bizkaia Health Research Institute, Barakaldo, Spain
- Department of Nursery, University of the Basque Country UPV/EHU, Leioa, Spain
| | - Inés Ibarra-Lecue
- Department of Pharmacology, University of the Basque Country, UPV/EHU, Leioa, Spain
| | - Igor Horrillo
- Department of Pharmacology, University of the Basque Country, UPV/EHU, Leioa, Spain
- Biocruces Bizkaia Health Research Institute, Barakaldo, Spain
- Centro de Investigación Biomédica en Red de Salud Mental, CIBERSAM, Madrid, Spain
| | - Aitor Villate
- Department of Analytical Chemistry, University of the Basque Country UPV/EHU, Leioa, Spain
- PiE-UPV/EHU, Plentzia, ItsasEstazioa, Areatza Pasealekua, 48620, Plentzia, Spain
| | - Maria Recio
- Biocruces Bizkaia Health Research Institute, Barakaldo, Spain
- Department of Psychiatry, Cruces University Hospital, Barakaldo, Spain
| | - J Javier Meana
- Department of Pharmacology, University of the Basque Country, UPV/EHU, Leioa, Spain
- Biocruces Bizkaia Health Research Institute, Barakaldo, Spain
- Centro de Investigación Biomédica en Red de Salud Mental, CIBERSAM, Madrid, Spain
| | - Rebeca Diez-Alarcia
- Department of Pharmacology, University of the Basque Country, UPV/EHU, Leioa, Spain
- Biocruces Bizkaia Health Research Institute, Barakaldo, Spain
- Centro de Investigación Biomédica en Red de Salud Mental, CIBERSAM, Madrid, Spain
| | - Oihane Mentxaka
- Biocruces Bizkaia Health Research Institute, Barakaldo, Spain
- Department of Psychiatry, Cruces University Hospital, Barakaldo, Spain
- Department of Neurosciences, University of the Basque Country, UPV/EHU, Leioa, Spain
| | - Rafael Segarra
- Biocruces Bizkaia Health Research Institute, Barakaldo, Spain
- Centro de Investigación Biomédica en Red de Salud Mental, CIBERSAM, Madrid, Spain
- Department of Psychiatry, Cruces University Hospital, Barakaldo, Spain
- Department of Neurosciences, University of the Basque Country, UPV/EHU, Leioa, Spain
| | - Nestor Etxebarria
- Department of Analytical Chemistry, University of the Basque Country UPV/EHU, Leioa, Spain
- PiE-UPV/EHU, Plentzia, ItsasEstazioa, Areatza Pasealekua, 48620, Plentzia, Spain
| | - Luis F Callado
- Department of Pharmacology, University of the Basque Country, UPV/EHU, Leioa, Spain
- Biocruces Bizkaia Health Research Institute, Barakaldo, Spain
- Centro de Investigación Biomédica en Red de Salud Mental, CIBERSAM, Madrid, Spain
| | - Leyre Urigüen
- Department of Pharmacology, University of the Basque Country, UPV/EHU, Leioa, Spain.
- Biocruces Bizkaia Health Research Institute, Barakaldo, Spain.
- Centro de Investigación Biomédica en Red de Salud Mental, CIBERSAM, Madrid, Spain.
| |
Collapse
|
5
|
Kostes WW, Brafman DA. The Multifaceted Role of WNT Signaling in Alzheimer's Disease Onset and Age-Related Progression. Cells 2023; 12:1204. [PMID: 37190113 PMCID: PMC10136584 DOI: 10.3390/cells12081204] [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: 03/05/2023] [Revised: 04/12/2023] [Accepted: 04/17/2023] [Indexed: 05/17/2023] Open
Abstract
The evolutionary conserved WNT signaling pathway orchestrates numerous complex biological processes during development and is critical to the maintenance of tissue integrity and homeostasis in the adult. As it relates to the central nervous system, WNT signaling plays several roles as it relates to neurogenesis, synaptic formation, memory, and learning. Thus, dysfunction of this pathway is associated with multiple diseases and disorders, including several neurodegenerative disorders. Alzheimer's disease (AD) is characterized by several pathologies, synaptic dysfunction, and cognitive decline. In this review, we will discuss the various epidemiological, clinical, and animal studies that demonstrate a precise link between aberrant WNT signaling and AD-associated pathologies. In turn, we will discuss the manner in which WNT signaling influences multiple molecular, biochemical, and cellular pathways upstream of these end-point pathologies. Finally, we will discuss how merging tools and technologies can be used to generate next generation cellular models to dissect the relationship between WNT signaling and AD.
Collapse
Affiliation(s)
| | - David A. Brafman
- School of Biological and Health Systems Engineering, Arizona State University, Tempe, AZ 85287, USA
| |
Collapse
|
6
|
Hong Y, Dong X, Chang L, Xie C, Chang M, Aguilar JS, Lin J, Lin J, Li QQ. Microglia-containing cerebral organoids derived from induced pluripotent stem cells for the study of neurological diseases. iScience 2023; 26:106267. [PMID: 36936782 PMCID: PMC10014280 DOI: 10.1016/j.isci.2023.106267] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2022] [Revised: 11/05/2022] [Accepted: 02/18/2023] [Indexed: 02/26/2023] Open
Abstract
Microglia play an important role in neuroinflammation and neurodegeneration. Here, we report an approach for generating microglia-containing cerebral organoids derived from human pluripotent stem cells involving the supplementation of growth factors (FGF, EGF, heparin) and 10% CO2 culture conditions. Using this platform, Western Pacific Amyotrophic Lateral Sclerosis and Parkinsonism-Dementia Complex (ALS-PDC) cerebral organoids were generated from patient-derived induced pluripotent stem cells (iPSCs). These ALS-PDC-affected organoids had more reactive astrocytes and M1 microglia, and had fewer M2 microglia than their unaffected counterparts, leading to impaired microglia-mediated phagocytosis. RNA-seq analysis of ALS-PDC and control organoids indicated that the most significant changes were microglia- and astrocyte-related genes (IFITM1/2, TGF-β, and GFAP). The most significantly downregulated pathway was type I interferon signaling. Interferon-gamma supplementation increased IFITM expression, enhanced microglia-mediated phagocytosis, and reduced beta-amyloid accumulation in ALS-PDC-affected network. The results demonstrated the feasibility of using microglia-containing organoids for the study of neurodegenerative diseases.
Collapse
Affiliation(s)
- Yiling Hong
- College of Veterinary Medicine, Western University of Health Sciences, Pomona, CA 91766-1854, USA
- Corresponding author
| | - Xu Dong
- Graduate College of Biomedical Sciences, Western University of Health Sciences, Pomona, CA 91766-1854, USA
| | - Lawrence Chang
- Graduate College of Biomedical Sciences, Western University of Health Sciences, Pomona, CA 91766-1854, USA
| | - Chen Xie
- College of Veterinary Medicine, Western University of Health Sciences, Pomona, CA 91766-1854, USA
| | - Mariann Chang
- College of Veterinary Medicine, Western University of Health Sciences, Pomona, CA 91766-1854, USA
| | - Jose S. Aguilar
- College of Veterinary Medicine, Western University of Health Sciences, Pomona, CA 91766-1854, USA
| | - Jimmy Lin
- Graduate College of Biomedical Sciences, Western University of Health Sciences, Pomona, CA 91766-1854, USA
| | - Juncheng Lin
- Graduate College of Biomedical Sciences, Western University of Health Sciences, Pomona, CA 91766-1854, USA
| | - Qingshun Q. Li
- Graduate College of Biomedical Sciences, Western University of Health Sciences, Pomona, CA 91766-1854, USA
- Biomedical Sciences, College of Dental Medicine, Western University of Health Sciences, Pomona, CA 91766-1854, USA
| |
Collapse
|
7
|
Transition from Animal-Based to Human Induced Pluripotent Stem Cells (iPSCs)-Based Models of Neurodevelopmental Disorders: Opportunities and Challenges. Cells 2023; 12:cells12040538. [PMID: 36831205 PMCID: PMC9954744 DOI: 10.3390/cells12040538] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Revised: 01/25/2023] [Accepted: 02/02/2023] [Indexed: 02/11/2023] Open
Abstract
Neurodevelopmental disorders (NDDs) arise from the disruption of highly coordinated mechanisms underlying brain development, which results in impaired sensory, motor and/or cognitive functions. Although rodent models have offered very relevant insights to the field, the translation of findings to clinics, particularly regarding therapeutic approaches for these diseases, remains challenging. Part of the explanation for this failure may be the genetic differences-some targets not being conserved between species-and, most importantly, the differences in regulation of gene expression. This prompts the use of human-derived models to study NDDS. The generation of human induced pluripotent stem cells (hIPSCs) added a new suitable alternative to overcome species limitations, allowing for the study of human neuronal development while maintaining the genetic background of the donor patient. Several hIPSC models of NDDs already proved their worth by mimicking several pathological phenotypes found in humans. In this review, we highlight the utility of hIPSCs to pave new paths for NDD research and development of new therapeutic tools, summarize the challenges and advances of hIPSC-culture and neuronal differentiation protocols and discuss the best way to take advantage of these models, illustrating this with examples of success for some NDDs.
Collapse
|
8
|
Whye D, Wood D, Saber WA, Norabuena EM, Makhortova NR, Sahin M, Buttermore ED. A Robust Pipeline for the Multi-Stage Accelerated Differentiation of Functional 3D Cortical Organoids from Human Pluripotent Stem Cells. Curr Protoc 2023; 3:e641. [PMID: 36633423 PMCID: PMC9839317 DOI: 10.1002/cpz1.641] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Disordered cellular development, abnormal neuroanatomical formations, and dysfunction of neuronal circuitry are among the pathological manifestations of cortical regions in the brain that are often implicated in complex neurodevelopmental disorders. With the advancement of stem cell methodologies such as cerebral organoid generation, it is possible to study these processes in vitro using 3D cellular platforms that mirror key developmental stages occurring throughout embryonic neurogenesis. Patterning-based stem cell models of directed neuronal development offer one approach to accomplish this, but these protocols often require protracted periods of cell culture to generate diverse cell types and current methods are plagued by a lack of specificity, reproducibility, and temporal control over cell derivation. Although ectopic expression of transcription factors offers another avenue to rapidly generate neurons, this process of direct lineage conversion bypasses critical junctures of neurodevelopment during which disease-relevant manifestations may occur. Here, we present a directed differentiation approach for generating human pluripotent stem cell (hPSC)-derived cortical organoids with accelerated lineage specification to generate functionally mature cortical neurons in a shorter timeline than previously established protocols. This novel protocol provides precise guidance for the specification of neuronal cell type identity as well as temporal control over the pace at which cortical lineage trajectories are established. Furthermore, we present assays that can be used as tools to interrogate stage-specific developmental signaling mechanisms. By recapitulating major components of embryonic neurogenesis, this protocol allows for improved in vitro modeling of cortical development while providing a platform that can be utilized to uncover disease-specific mechanisms of disordered development at various stages across the differentiation timeline. © 2023 Wiley Periodicals LLC. Basic Protocol 1: 3D hPSC neural induction Support Protocol 1: Neural rosette formation assay Support Protocol 2: Neurosphere generation Support Protocol 3: Enzymatic dissociation, NSC expansion, and cryopreservation Basic Protocol 2: 3D neural progenitor expansion Basic Protocol 3: 3D accelerated cortical lineage patterning and terminal differentiation.
Collapse
Affiliation(s)
- Dosh Whye
- Human Neuron Core, Rosamund Stone Zander Translational Neuroscience Center, Boston Children’s Hospital, Boston, MA
- F.M. Kirby Neurobiology Department, Boston Children’s Hospital, Boston, MA
| | - Delaney Wood
- Human Neuron Core, Rosamund Stone Zander Translational Neuroscience Center, Boston Children’s Hospital, Boston, MA
- F.M. Kirby Neurobiology Department, Boston Children’s Hospital, Boston, MA
| | - Wardiya Afshar Saber
- Human Neuron Core, Rosamund Stone Zander Translational Neuroscience Center, Boston Children’s Hospital, Boston, MA
- F.M. Kirby Neurobiology Department, Boston Children’s Hospital, Boston, MA
- Department of Neurology, Harvard Medical School, Boston, MA
| | - Erika M. Norabuena
- Human Neuron Core, Rosamund Stone Zander Translational Neuroscience Center, Boston Children’s Hospital, Boston, MA
- F.M. Kirby Neurobiology Department, Boston Children’s Hospital, Boston, MA
| | - Nina R. Makhortova
- Human Neuron Core, Rosamund Stone Zander Translational Neuroscience Center, Boston Children’s Hospital, Boston, MA
- F.M. Kirby Neurobiology Department, Boston Children’s Hospital, Boston, MA
- Department of Neurology, Harvard Medical School, Boston, MA
| | - Mustafa Sahin
- Human Neuron Core, Rosamund Stone Zander Translational Neuroscience Center, Boston Children’s Hospital, Boston, MA
- F.M. Kirby Neurobiology Department, Boston Children’s Hospital, Boston, MA
- Department of Neurology, Harvard Medical School, Boston, MA
| | - Elizabeth D. Buttermore
- Human Neuron Core, Rosamund Stone Zander Translational Neuroscience Center, Boston Children’s Hospital, Boston, MA
- F.M. Kirby Neurobiology Department, Boston Children’s Hospital, Boston, MA
| |
Collapse
|
9
|
Schaefers C, Rothmiller S, Thiermann H, Rein T, Schmidt A. The Efficiency of Direct Maturation: the Comparison of Two hiPSC Differentiation Approaches into Motor Neurons. Stem Cells Int 2022; 2022:1320950. [PMID: 36530489 PMCID: PMC9757946 DOI: 10.1155/2022/1320950] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2022] [Revised: 11/10/2022] [Accepted: 11/11/2022] [Indexed: 02/23/2025] Open
Abstract
Motor neurons (MNs) derived from human-induced pluripotent stem cells (hiPSC) hold great potential for the treatment of various motor neurodegenerative diseases as transplantations with a low-risk of rejection are made possible. There are many hiPSC differentiation protocols that pursue to imitate the multistep process of motor neurogenesis in vivo. However, these often apply viral vectors, feeder cells, or antibiotics to generate hiPSC and MNs, limiting their translational potential. In this study, a virus-, feeder-, and antibiotic-free method was used for reprogramming hiPSC, which were maintained in culture medium produced under clinical good manufacturing practice. Differentiation into MNs was performed with standardized, chemically defined, and antibiotic-free culture media. The identity of hiPSC, neuronal progenitors, and mature MNs was continuously verified by the detection of specific markers at the genetic and protein level via qRT-PCR, flow cytometry, Western Blot, and immunofluorescence. MNX1- and ChAT-positive motoneuronal progenitor cells were formed after neural induction via dual-SMAD inhibition and expansion. For maturation, an approach aiming to directly mature these progenitors was compared to an approach that included an additional differentiation step for further specification. Although both approaches generated mature MNs expressing characteristic postmitotic markers, the direct maturation approach appeared to be more efficient. These results provide new insights into the suitability of two standardized differentiation approaches for generating mature MNs, which might pave the way for future clinical applications.
Collapse
Affiliation(s)
- Catherine Schaefers
- Bundeswehr Institute of Pharmacology and Toxicology, Neuherbergstr. 11, 80937 Munich, Germany
| | - Simone Rothmiller
- Bundeswehr Institute of Pharmacology and Toxicology, Neuherbergstr. 11, 80937 Munich, Germany
| | - Horst Thiermann
- Bundeswehr Institute of Pharmacology and Toxicology, Neuherbergstr. 11, 80937 Munich, Germany
| | - Theo Rein
- Max Planck Institute of Psychiatry, Kraepelinstr. 2-10, 80804 Munich, Germany
| | - Annette Schmidt
- Bundeswehr Institute of Pharmacology and Toxicology, Neuherbergstr. 11, 80937 Munich, Germany
- Institute of Sport Science, University of the Bundeswehr Munich, Werner-Heisenberg-Weg 39, 85577 Neubiberg, Germany
| |
Collapse
|
10
|
Li H, Li QQ, Hong Y. Global gene expression signatures in response to citrate-coated silver nanoparticles exposure. Toxicology 2021; 461:152898. [PMID: 34403730 DOI: 10.1016/j.tox.2021.152898] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2021] [Revised: 08/04/2021] [Accepted: 08/12/2021] [Indexed: 10/20/2022]
Abstract
Silver nanoparticles (AgNPs) are widely used in medical and commercial products for their unique antibacterial functions. However, the impact of AgNPs on human neural development is not well understood. To investigate the effect of AgNPs on human neural development, various doses of 20 nm citrate-coated AgNP (AgSC) were administered to human embryonic stem cell derived neural progenitors during the neuronal differentiation. Immunofluorescence staining with neuronal progenitor markers SOX2 (sex determining region Y-box 2) and Nestin (VI intermediate filament protein) showed that AgSC inhibited rosette formation, neuronal progenitor proliferation, and neurite outgrowth. Furthermore, AgSC promoted astrocyte activation and neuronal apoptosis. These adverse effects can be partially recovered with ascorbic acid. A genome-wide transcriptome analysis of both AgSC treated and untreated samples indicated that the most up-graduated genes were a group of Metallothionein (1F, 1E, 2A) proteins, a metal-binding protein that plays an essential role in metal homeostasis, heavy metal detoxification, and cellular anti-oxidative defence. The most significantly down-regulated genes were neuronal differentiation 6 (NEUROD6) and fork head box G1 (FOXG1). GO analyse indicated that the regulation of cholesterol biosynthetic process, neuron differentiation, synapse organization and pattern specification, oliogenesis, and neuronal apoptosis were the most impacted biological processes. KEGG pathway analyse showed that the most significantly impacted pathways were C5 isoprenoid, axon guidance, Notch, WNT, RAS-MAPK signalling pathways, lysosome, and apoptosis. Our data suggests that AgSCs interfered with metal homeostasis and cholesterol biosynthesis which induced oxidative stress, inhibited neurogenesis, axon guidance, and promoted apoptosis. Supplementation with ascorbic acid could act as an antioxidant to prevent AgSC-mediated neurotoxicity.
Collapse
Affiliation(s)
- Hao Li
- Key Laboratory of the Ministry of Education for Coastal and Wetland Ecosystem, College of the Environment and Ecology, Xiamen University, Xiamen, Fujian, 361102, China; Graduate College of Biomedical Sciences, Western University of Health Sciences, Pomona, CA, 91766-1854, USA
| | - Qingshun Q Li
- Key Laboratory of the Ministry of Education for Coastal and Wetland Ecosystem, College of the Environment and Ecology, Xiamen University, Xiamen, Fujian, 361102, China; Graduate College of Biomedical Sciences, Western University of Health Sciences, Pomona, CA, 91766-1854, USA.
| | - Yiling Hong
- College of Veterinary Medicine, Western University of Health Sciences, Pomona, CA, 91766-1854, USA.
| |
Collapse
|
11
|
Characterisation of Neurospheres-Derived Cells from Human Olfactory Epithelium. Cells 2021; 10:cells10071690. [PMID: 34359860 PMCID: PMC8307784 DOI: 10.3390/cells10071690] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 06/25/2021] [Accepted: 06/30/2021] [Indexed: 11/17/2022] Open
Abstract
A major problem in psychiatric research is a deficit of relevant cell material of neuronal origin, especially in large quantities from living individuals. One of the promising options is cells from the olfactory neuroepithelium, which contains neuronal progenitors that ensure the regeneration of olfactory receptors. These cells are easy to obtain with nasal biopsies and it is possible to grow and cultivate them in vitro. In this work, we used RNAseq expression profiling and immunofluorescence microscopy to characterise neurospheres-derived cells (NDC), that simply and reliably grow from neurospheres (NS) obtained from nasal biopsies. We utilized differential expression analysis to explore the molecular changes that occur during transition from NS to NDC. We found that processes associated with neuronal and vascular cells are downregulated in NDC. A comparison with public transcriptomes revealed a depletion of neuronal and glial components in NDC. We also discovered that NDC have several metabolic features specific to neuronal progenitors treated with the fungicide maneb. Thus, while NDC retain some neuronal/glial identity, additional protocol alterations are needed to use NDC for mass sample collection in psychiatric research.
Collapse
|
12
|
Oyefeso FA, Muotri AR, Wilson CG, Pecaut MJ. Brain organoids: A promising model to assess oxidative stress-induced central nervous system damage. Dev Neurobiol 2021; 81:653-670. [PMID: 33942547 DOI: 10.1002/dneu.22828] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 04/28/2021] [Accepted: 04/29/2021] [Indexed: 12/13/2022]
Abstract
Oxidative stress (OS) is one of the most significant propagators of systemic damage with implications for widespread pathologies such as vascular disease, accelerated aging, degenerative disease, inflammation, and traumatic injury. OS can be induced by numerous factors such as environmental conditions, lifestyle choices, disease states, and genetic susceptibility. It is tied to the accumulation of free radicals, mitochondrial dysfunction, and insufficient antioxidant protection, which leads to cell aging and tissue degeneration over time. Unregulated systemic increase in reactive species, which contain harmful free radicals, can lead to diverse tissue-specific OS responses and disease. Studies of OS in the brain, for example, have demonstrated how this state contributes to neurodegeneration and altered neural plasticity. As the worldwide life expectancy has increased over the last few decades, the prevalence of OS-related diseases resulting from age-associated progressive tissue degeneration. Unfortunately, vital translational research studies designed to identify and target disease biomarkers in human patients have been impeded by many factors (e.g., limited access to human brain tissue for research purposes and poor translation of experimental models). In recent years, stem cell-derived three-dimensional tissue cultures known as "brain organoids" have taken the spotlight as a novel model for studying central nervous system (CNS) diseases. In this review, we discuss the potential of brain organoids to model the responses of human neural cells to OS, noting current and prospective limitations. Overall, brain organoids show promise as an innovative translational model to study CNS susceptibility to OS and elucidate the pathophysiology of the aging brain.
Collapse
Affiliation(s)
- Foluwasomi A Oyefeso
- Department of Biomedical Engineering Sciences, School of Medicine, Loma Linda University, Loma Linda, CA, USA
| | - Alysson R Muotri
- Department of Pediatrics/Cellular and Molecular Medicine, University of California San Diego, San Diego, CA, USA
| | - Christopher G Wilson
- Lawrence D. Longo, MD, Center for Perinatal Biology, School of Medicine, Loma Linda University, Loma Linda, CA, USA
| | - Michael J Pecaut
- Department of Biomedical Engineering Sciences, School of Medicine, Loma Linda University, Loma Linda, CA, USA
| |
Collapse
|
13
|
Si Z, Wang X. Stem Cell Therapies in Alzheimer's Disease: Applications for Disease Modeling. J Pharmacol Exp Ther 2021; 377:207-217. [PMID: 33558427 DOI: 10.1124/jpet.120.000324] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Accepted: 02/03/2021] [Indexed: 12/19/2022] Open
Abstract
Alzheimer's disease (AD) is a neurodegenerative disease with complex pathologic and biologic characteristics. Extracellular β-amyloid deposits, such as senile plaques, and intracellular aggregation of hyperphosphorylated tau, such as neurofibrillary tangles, remain the main neuropathological criteria for the diagnosis of AD. There is currently no effective treatment of the disease, and many clinical trials have failed to prove any benefits of new therapeutics. More recently, there has been increasing interest in harnessing the potential of stem cell technologies for drug discovery, disease modeling, and cell therapies, which have been used to study an array of human conditions, including AD. The recently developed and optimized induced pluripotent stem cell (iPSC) technology is a critical platform for screening anti-AD drugs and understanding mutations that modify AD. Neural stem cell (NSC) transplantation has been investigated as a new therapeutic approach to treat neurodegenerative diseases. Mesenchymal stem cells (MSCs) also exhibit considerable potential to treat neurodegenerative diseases by secreting growth factors and exosomes, attenuating neuroinflammation. This review highlights recent progress in stem cell research and the translational applications and challenges of iPSCs, NSCs, and MSCs as treatment strategies for AD. Even though these treatments are still in relative infancy, these developing stem cell technologies hold considerable promise to combat AD and other neurodegenerative disorders. SIGNIFICANCE STATEMENT: Alzheimer's disease (AD) is a neurodegenerative disease that results in learning and memory defects. Although some drugs have been approved for AD treatment, fewer than 20% of patients with AD benefit from these drugs. Therapies based on stem cells, including induced pluripotent stem cells, neural stem cells, and mesenchymal stem cells, provide promising therapeutic strategies for AD.
Collapse
Affiliation(s)
- Zizhen Si
- Department of Physiology and Pharmacology, School of Medicine, Ningbo University, Ningbo, China (Z.S.) and Department of Biochemistry and Molecular Biology, Harbin Medical University, Harbin, China (X.W.)
| | - Xidi Wang
- Department of Physiology and Pharmacology, School of Medicine, Ningbo University, Ningbo, China (Z.S.) and Department of Biochemistry and Molecular Biology, Harbin Medical University, Harbin, China (X.W.)
| |
Collapse
|
14
|
MacDougall G, Brown LY, Kantor B, Chiba-Falek O. The Path to Progress Preclinical Studies of Age-Related Neurodegenerative Diseases: A Perspective on Rodent and hiPSC-Derived Models. Mol Ther 2021; 29:949-972. [PMID: 33429080 PMCID: PMC7934639 DOI: 10.1016/j.ymthe.2021.01.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Revised: 12/03/2020] [Accepted: 01/01/2021] [Indexed: 12/21/2022] Open
Abstract
Alzheimer's disease (AD) and Parkinson's disease (PD) are the two most prevalent age-related neurodegenerative diseases, and currently no effective clinical treatments exist for either, despite decades of clinical trials. The failure to translate preclinical findings into effective treatments is indicative of a problem in the current evaluation pipeline for potential therapeutics. At present, there are no useful animal models for AD and PD research that reflect the entire biology of the diseases, specifically, the more common non-Mendelian forms. Whereas the field continues to seek suitable rodent models for investigating potential therapeutics for these diseases, rodent models have still been used primarily for preclinical studies. Here, we advocate for a paradigm shift toward the application of human-induced pluripotent stem cell (hiPSC)-derived systems for PD and AD modeling and the development of improved human-based models in a dish for drug discovery and preclinical assessment of therapeutic targets.
Collapse
Affiliation(s)
- Gabriella MacDougall
- Division of Translational Brain Sciences, Department of Neurology, Duke University Medical Center, Durham, NC 27710, USA; Center for Genomic and Computational Biology, Duke University Medical Center, Durham, NC 27710, USA
| | - Logan Y Brown
- Department of Neurobiology, Duke University Medical Center, Durham, NC 27710, USA; Center for Advanced Genomic Technologies, Duke University Medical Center, Durham, NC 27710, USA; Viral Vector Core, Duke University Medical Center, Durham, NC 27710, USA
| | - Boris Kantor
- Department of Neurobiology, Duke University Medical Center, Durham, NC 27710, USA; Center for Advanced Genomic Technologies, Duke University Medical Center, Durham, NC 27710, USA; Viral Vector Core, Duke University Medical Center, Durham, NC 27710, USA.
| | - Ornit Chiba-Falek
- Division of Translational Brain Sciences, Department of Neurology, Duke University Medical Center, Durham, NC 27710, USA; Center for Genomic and Computational Biology, Duke University Medical Center, Durham, NC 27710, USA.
| |
Collapse
|
15
|
Identification of Molecular Signatures in Neural Differentiation and Neurological Diseases Using Digital Color-Coded Molecular Barcoding. Stem Cells Int 2020; 2020:8852313. [PMID: 33005195 PMCID: PMC7503121 DOI: 10.1155/2020/8852313] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Revised: 08/26/2020] [Accepted: 08/28/2020] [Indexed: 12/04/2022] Open
Abstract
Human pluripotent stem cells (PSCs), including embryonic stem cells and induced pluripotent stem cells, represent powerful tools for disease modeling and for therapeutic applications. PSCs are particularly useful for the study of development and diseases of the nervous system. However, generating in vitro models that recapitulate the architecture and the full variety of subtypes of cells that make the complexity of our brain remains a challenge. In order to fully exploit the potential of PSCs, advanced methods that facilitate the identification of molecular signatures in neural differentiation and neurological diseases are highly demanded. Here, we review the literature on the development and application of digital color-coded molecular barcoding as a potential tool for standardizing PSC research and applications in neuroscience. We will also describe relevant examples of the use of this technique for the characterization of the heterogeneous composition of the brain tumor glioblastoma multiforme.
Collapse
|
16
|
Raman S, Brookhouser N, Brafman DA. Using human induced pluripotent stem cells (hiPSCs) to investigate the mechanisms by which Apolipoprotein E (APOE) contributes to Alzheimer's disease (AD) risk. Neurobiol Dis 2020; 138:104788. [PMID: 32032733 PMCID: PMC7098264 DOI: 10.1016/j.nbd.2020.104788] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2019] [Revised: 01/14/2020] [Accepted: 01/31/2020] [Indexed: 01/02/2023] Open
Abstract
Although the biochemical and pathological hallmarks of Alzheimer's disease (AD), such as axonal transport defects, synaptic loss, and selective neuronal death, are well characterized, the underlying mechanisms that cause AD are largely unknown, thereby making it difficult to design effective therapeutic interventions. Genome-wide association studies (GWAS) studies have identified several factors associated with increased AD risk. Of these genetic factors, polymorphisms in the Apolipoprotein E (APOE) gene are the strongest and most prevalent. While it has been established that the ApoE protein modulates the formation of amyloid plaques and neurofibrillary tangles, the precise molecular mechanisms by which various ApoE isoforms enhance or mitigate AD onset and progression in aging adults are yet to be elucidated. Advances in cellular reprogramming to generate disease-in-a-dish models now provide a simplified and accessible system that complements animal and primary cell models to study ApoE in the context of AD. In this review, we will describe the use and manipulation of human induced pluripotent stem cells (hiPSCs) in dissecting the interaction between ApoE and AD. First, we will provide an overview of the proposed roles that ApoE plays in modulating pathophysiology of AD. Next, we will summarize the recent studies that have employed hiPSCs to model familial and sporadic AD. Lastly, we will speculate on how current advances in genome editing technologies and organoid culture systems can be used to improve hiPSC-based tools to investigate ApoE-dependent modulation of AD onset and progression.
Collapse
Affiliation(s)
- Sreedevi Raman
- School of Biological and Health Systems Engineering, Arizona State University, United States of America
| | - Nicholas Brookhouser
- School of Biological and Health Systems Engineering, Arizona State University, United States of America; Graduate Program in Clinical Translational Sciences, University of Arizona College of Medicine-Phoenix, United States of America
| | - David A Brafman
- School of Biological and Health Systems Engineering, Arizona State University, United States of America.
| |
Collapse
|
17
|
Penney J, Ralvenius WT, Tsai LH. Modeling Alzheimer's disease with iPSC-derived brain cells. Mol Psychiatry 2020; 25:148-167. [PMID: 31391546 PMCID: PMC6906186 DOI: 10.1038/s41380-019-0468-3] [Citation(s) in RCA: 274] [Impact Index Per Article: 54.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Revised: 04/10/2019] [Accepted: 05/13/2019] [Indexed: 12/21/2022]
Abstract
Alzheimer's disease is a devastating neurodegenerative disorder with no cure. Countless promising therapeutics have shown efficacy in rodent Alzheimer's disease models yet failed to benefit human patients. While hope remains that earlier intervention with existing therapeutics will improve outcomes, it is becoming increasingly clear that new approaches to understand and combat the pathophysiology of Alzheimer's disease are needed. Human induced pluripotent stem cell (iPSC) technologies have changed the face of preclinical research and iPSC-derived cell types are being utilized to study an array of human conditions, including neurodegenerative disease. All major brain cell types can now be differentiated from iPSCs, while increasingly complex co-culture systems are being developed to facilitate neuroscience research. Many cellular functions perturbed in Alzheimer's disease can be recapitulated using iPSC-derived cells in vitro, and co-culture platforms are beginning to yield insights into the complex interactions that occur between brain cell types during neurodegeneration. Further, iPSC-based systems and genome editing tools will be critical in understanding the roles of the numerous new genes and mutations found to modify Alzheimer's disease risk in the past decade. While still in their relative infancy, these developing iPSC-based technologies hold considerable promise to push forward efforts to combat Alzheimer's disease and other neurodegenerative disorders.
Collapse
Affiliation(s)
- Jay Penney
- Department of Brain and Cognitive Sciences, Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - William T Ralvenius
- Department of Brain and Cognitive Sciences, Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Li-Huei Tsai
- Department of Brain and Cognitive Sciences, Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA.
| |
Collapse
|
18
|
Wei WJ, Shi B, Guan X, Ma JY, Wang YC, Liu J. Mapping theme trends and knowledge structures for human neural stem cells: a quantitative and co-word biclustering analysis for the 2013-2018 period. Neural Regen Res 2019; 14:1823-1832. [PMID: 31169201 PMCID: PMC6585554 DOI: 10.4103/1673-5374.257535] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2018] [Accepted: 03/06/2019] [Indexed: 01/27/2023] Open
Abstract
Neural stem cells, which are capable of multi-potential differentiation and self-renewal, have recently been shown to have clinical potential for repairing central nervous system tissue damage. However, the theme trends and knowledge structures for human neural stem cells have not yet been studied bibliometrically. In this study, we retrieved 2742 articles from the PubMed database from 2013 to 2018 using "Neural Stem Cells" as the retrieval word. Co-word analysis was conducted to statistically quantify the characteristics and popular themes of human neural stem cell-related studies. Bibliographic data matrices were generated with the Bibliographic Item Co-Occurrence Matrix Builder. We identified 78 high-frequency Medical Subject Heading (MeSH) terms. A visual matrix was built with the repeated bisection method in gCLUTO software. A social network analysis network was generated with Ucinet 6.0 software and GraphPad Prism 5 software. The analyses demonstrated that in the 6-year period, hot topics were clustered into five categories. As suggested by the constructed strategic diagram, studies related to cytology and physiology were well-developed, whereas those related to neural stem cell applications, tissue engineering, metabolism and cell signaling, and neural stem cell pathology and virology remained immature. Neural stem cell therapy for stroke and Parkinson's disease, the genetics of microRNAs and brain neoplasms, as well as neuroprotective agents, Zika virus, Notch receptor, neural crest and embryonic stem cells were identified as emerging hot spots. These undeveloped themes and popular topics are potential points of focus for new studies on human neural stem cells.
Collapse
Affiliation(s)
- Wen-Juan Wei
- Stem Cell Clinical Research Center, the First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning Province, China
- National Joint Engineering Laboratory, the First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning Province, China
| | - Bei Shi
- Department of Physiology, China Medical University, Shenyang, Liaoning Province, China
| | - Xin Guan
- Stem Cell Clinical Research Center, the First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning Province, China
- National Joint Engineering Laboratory, the First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning Province, China
| | - Jing-Yun Ma
- Stem Cell Clinical Research Center, the First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning Province, China
- National Joint Engineering Laboratory, the First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning Province, China
| | - Ya-Chen Wang
- Stem Cell Clinical Research Center, the First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning Province, China
- National Joint Engineering Laboratory, the First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning Province, China
| | - Jing Liu
- Stem Cell Clinical Research Center, the First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning Province, China
- National Joint Engineering Laboratory, the First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning Province, China
| |
Collapse
|
19
|
Wang H, Chang L, Aguilar JS, Dong S, Hong Y. Bisphenol-A exposure induced neurotoxicity in glutamatergic neurons derived from human embryonic stem cells. ENVIRONMENT INTERNATIONAL 2019; 127:324-332. [PMID: 30953815 DOI: 10.1016/j.envint.2019.01.059] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/05/2018] [Revised: 01/22/2019] [Accepted: 01/22/2019] [Indexed: 06/09/2023]
Abstract
Bisphenol-A (BPA) is a lipophilic, organic, synthetic compound that has been used as an additive in polycarbonate plastics manufacturing since 1957. Studies have shown that BPA interferes with the development and functions of the brain, but little is known about the effects of BPA on human glutamatergic neurons (hGNs) at the molecular and cellular levels. We investigated the impact of chronic exposure to BPA to hGNs derived from human embryonic stem cells (hESCs). The results showed that chronic exposure of different concentrations of BPA (0, 0.1, 1.0 and 10 μM) to hGNs for 14 days reduced neurite outgrowth in a concentration-dependent manner. Using presynaptic protein synaptophysin and postsynaptic protein PSD-95 antibodies, immunofluorescence staining and western blotting results indicated that BPA exposure altered the morphology of dendritic spines and increased synaptophysin and PSD-95 expression. Furthermore, BPA exposure at concentrations higher than 1.0 μM resulted in the increase of α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor (AMPAR) expression and deterioration of dendritic spines. In addition, our results suggested that these BPA mediated neurotoxicity effects were due to an increased production of reactive nitrogen species (RNS) and reactive oxygen species (ROS) via increased nitric oxide synthase (iNOS), neuronal nitric oxide synthase (nNOS), 3-nitrotyrosine expression and Ca2+ influx. These results imply that hESC-based neuronal differentiation is an excellent cellular model to examine BPA-induced neurotoxicity on human neurons at the cellular and molecular level.
Collapse
Affiliation(s)
- Hongou Wang
- College of Veterinary Medicine, Western University of Health Sciences, Pomona, CA 91766-1854, USA; Center for Excellence in Regional Atmospheric Environment, Institute of Urban Environment, Chinese Academy of Sciences, Xiamen, 361021, China; Key Lab of Urban Environment and Health, Institute of Urban Environment, Chinese Academy of Sciences, Xiamen, 361021, China; College of Resources and Environment, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Lawrence Chang
- College of Veterinary Medicine, Western University of Health Sciences, Pomona, CA 91766-1854, USA; Graduate College of Biomedical Sciences, Western University of Health Sciences, Pomona, CA 91766-1854, USA
| | - Jose S Aguilar
- Graduate College of Biomedical Sciences, Western University of Health Sciences, Pomona, CA 91766-1854, USA
| | - Sijun Dong
- Center for Excellence in Regional Atmospheric Environment, Institute of Urban Environment, Chinese Academy of Sciences, Xiamen, 361021, China; Key Lab of Urban Environment and Health, Institute of Urban Environment, Chinese Academy of Sciences, Xiamen, 361021, China.
| | - Yiling Hong
- College of Veterinary Medicine, Western University of Health Sciences, Pomona, CA 91766-1854, USA; Graduate College of Biomedical Sciences, Western University of Health Sciences, Pomona, CA 91766-1854, USA.
| |
Collapse
|
20
|
Iwashita M, Ohta H, Fujisawa T, Cho M, Ikeya M, Kidoaki S, Kosodo Y. Brain-stiffness-mimicking tilapia collagen gel promotes the induction of dorsal cortical neurons from human pluripotent stem cells. Sci Rep 2019; 9:3068. [PMID: 30816128 PMCID: PMC6395773 DOI: 10.1038/s41598-018-38395-5] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2018] [Accepted: 12/21/2018] [Indexed: 11/08/2022] Open
Abstract
The mechanical properties of the extracellular microenvironment, including its stiffness, play a crucial role in stem cell fate determination. Although previous studies have demonstrated that the developing brain exhibits spatiotemporal diversity in stiffness, it remains unclear how stiffness regulates stem cell fate towards specific neural lineages. Here, we established a culture substrate that reproduces the stiffness of brain tissue using tilapia collagen for in vitro reconstitution assays. By adding crosslinkers, we obtained gels that are similar in stiffness to living brain tissue (150-1500 Pa). We further examined the capability of the gels serving as a substrate for stem cell culture and the effect of stiffness on neural lineage differentiation using human iPS cells. Surprisingly, exposure to gels with a stiffness of approximately 1500 Pa during the early period of neural induction promoted the production of dorsal cortical neurons. These findings suggest that brain-stiffness-mimicking gel has the potential to determine the terminal neural subtype. Taken together, the crosslinked tilapia collagen gel is expected to be useful in various reconstitution assays that can be used to explore the role of stiffness in neurogenesis and neural functions. The enhanced production of dorsal cortical neurons may also provide considerable advantages for neural regenerative applications.
Collapse
Affiliation(s)
- Misato Iwashita
- Korea Brain Research Institute, 61, Chemdan-ro, Dong-gu, Daegu, 41068, Republic of Korea
| | - Hatsumi Ohta
- Ihara & Co, Ltd, 3-263-23, Zenibako, Otaru, Hokkaido, 947-0261, Japan
| | - Takahiro Fujisawa
- Institute for Materials Chemistry and Engineering, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka, 819-0395, Japan
| | - Minyoung Cho
- Korea Brain Research Institute, 61, Chemdan-ro, Dong-gu, Daegu, 41068, Republic of Korea
| | - Makoto Ikeya
- Center for iPS Cell Research and Application (CiRA), Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Satoru Kidoaki
- Institute for Materials Chemistry and Engineering, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka, 819-0395, Japan
| | - Yoichi Kosodo
- Korea Brain Research Institute, 61, Chemdan-ro, Dong-gu, Daegu, 41068, Republic of Korea.
| |
Collapse
|
21
|
Hong Y, Chan N, Begum AN. Deriving Neural Cells from Pluripotent Stem Cells for Nanotoxicity Testing. Methods Mol Biol 2019; 1894:57-72. [PMID: 30547455 DOI: 10.1007/978-1-4939-8916-4_4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Stem cells are undifferentiated biological cells that can differentiate into all lineages under defined control condition. Stem cell neuronal differentiation can faithfully recapitulate stages of neural development and generate neuronal progenitors, mature neurons, and glial cells. Stem cell technology will largely allow for the replacement of animal studies and reduce costs, and will provide a new paradigm for in toxic genomics, bioinformatics, systems biology, and epigenetics studies. Here, we describe a nonadherent neuronal differentiation methodology developed in our laboratory, which can rapidly derive neurons and astrocytes from human embryonic stem cells (hESCs) and induced pluripotent stem cell (hiPSC) and use of this platform for nanoparticle neurotoxicity study.
Collapse
Affiliation(s)
- Yiling Hong
- College of Veterinary Medicine, Western University of Health Sciences, Pomona, CA, USA.
| | - Nymph Chan
- College of Veterinary Medicine, Western University of Health Sciences, Pomona, CA, USA
| | - Aynun N Begum
- College of Veterinary Medicine, Western University of Health Sciences, Pomona, CA, USA
| |
Collapse
|
22
|
Repar N, Li H, Aguilar JS, Li QQ, Damjana D, Hong Y. Silver nanoparticles induce neurotoxicity in a human embryonic stem cell-derived neuron and astrocyte network. Nanotoxicology 2018; 12:104-116. [PMID: 29334833 PMCID: PMC6172039 DOI: 10.1080/17435390.2018.1425497] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2017] [Revised: 01/05/2018] [Accepted: 01/05/2018] [Indexed: 10/18/2022]
Abstract
Silver nanoparticles (AgNPs) are among the most extensively used nanoparticles and are found in a variety of products. This ubiquity leads to inevitable exposure to these particles in everyday life. However, the effects of AgNPs on neuron and astrocyte networks are still largely unknown. In this study, we used neurons and astrocytes derived from human embryonic stem cells as a cellular model to study the neurotoxicity that is induced by citrate-coated AgNPs (AgSCs). Immunostaining with the astrocyte and neuron markers, glial fibrillary acidic protein and microtubule-associated protein-2 (MAP2), respectively, showed that exposure to AgSCs at the concentration of 0.1 µg/mL increased the astrocyte/neuron ratio. In contrast, a higher concentration of AgSCs (5.0 µg/ml) significantly changed the morphology of astrocytes. These results suggest that astrocytes are sensitive to AgSC exposure and that low concentrations of AgSCs promote astrogenesis. Furthermore, our results showed that AgSCs reduced neurite outgrowth, decreased the expression of postsynaptic density protein 95 and synaptophysin, and induced neurodegeneration in a concentration-dependent manner. Our findings additionally suggest that the expression and phosphorylation status of MAP2 isoforms, as modulated by the activation of the Akt/glycogen synthase kinase-3/caspase-3 signaling pathway, may play an important role in AgSC-mediated neurotoxicity. We also found that AgNO3 exposure only slightly reduced neurite outgrowth and had little effect on MAP2 expression, suggesting that AgSCs and AgNO3 have different neuronal toxicity mechanisms. In addition, most of these effects were reduced when the cell culture was co-treated with AgSCs and the antioxidant ascorbic acid, which implies that oxidative stress is the major cause of AgSC-mediated astrocytic/neuronal toxicity and that antioxidants may have a neuroprotective effect.
Collapse
Affiliation(s)
- Neza Repar
- College of Veterinary Medicine, Western University of Health Sciences, Pomona, CA 91766-1854, USA
- Department of Biology, Biotechnical Faculty, University of Ljubljana, Vecna pot 111, 1000 Ljubljana, Slovenia
| | - Hao Li
- Graduate College of Biomedical Sciences, Western University of Health Sciences, Pomona, CA 91766-1854, USA
- College of the Environment and Ecology, Xiamen University, Xiamen, Fujian 361002, China
| | - Jose S. Aguilar
- College of Veterinary Medicine, Western University of Health Sciences, Pomona, CA 91766-1854, USA
| | - Qingshun Q. Li
- Graduate College of Biomedical Sciences, Western University of Health Sciences, Pomona, CA 91766-1854, USA
- College of the Environment and Ecology, Xiamen University, Xiamen, Fujian 361002, China
| | - Drobne Damjana
- Department of Biology, Biotechnical Faculty, University of Ljubljana, Vecna pot 111, 1000 Ljubljana, Slovenia
| | - Yiling Hong
- College of Veterinary Medicine, Western University of Health Sciences, Pomona, CA 91766-1854, USA
| |
Collapse
|
23
|
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: 101] [Impact Index Per Article: 14.4] [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
|
24
|
McMurtrey RJ. Roles of Diffusion Dynamics in Stem Cell Signaling and Three-Dimensional Tissue Development. Stem Cells Dev 2017; 26:1293-1303. [PMID: 28707964 PMCID: PMC5610402 DOI: 10.1089/scd.2017.0066] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2017] [Accepted: 07/13/2017] [Indexed: 12/13/2022] Open
Abstract
Recent advancements in the ability to construct three-dimensional (3D) tissues and organoids from stem cells and biomaterials have not only opened abundant new research avenues in disease modeling and regenerative medicine but also have ignited investigation into important aspects of molecular diffusion in 3D cellular architectures. This article describes fundamental mechanics of diffusion with equations for modeling these dynamic processes under a variety of scenarios in 3D cellular tissue constructs. The effects of these diffusion processes and resultant concentration gradients are described in the context of the major molecular signaling pathways in stem cells that both mediate and are influenced by gas and nutrient concentrations, including how diffusion phenomena can affect stem cell state, cell differentiation, and metabolic states of the cell. The application of these diffusion models and pathways is of vital importance for future studies of developmental processes, disease modeling, and tissue regeneration.
Collapse
|
25
|
Hříbková H, Zelinková J, Sun YM. Progress in human pluripotent stem cell-based modeling systems for neurological diseases. NEUROGENESIS 2017. [DOI: 10.1080/23262133.2017.1324258] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Affiliation(s)
- Hana Hříbková
- Department of Biology, Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Jana Zelinková
- Department of Biology, Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Yuh-Man Sun
- Department of Biology, Faculty of Medicine, Masaryk University, Brno, Czech Republic
| |
Collapse
|
26
|
Adelaja AA. Human induced pluripotent stem cells generated neural cells behaving like brain and spinal cord cells: An insight into the involvement of retinoic acid and sonic hedgehog proteins. Int J Health Sci (Qassim) 2017; 11:21-27. [PMID: 28539859 PMCID: PMC5426416] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
OBJECTIVES The previous studies generated neural progenitor cells (NPCs) from human induced pluripotent stem cells (hiPSCs) using different protocols. However, the nature of the temporal or regional specificity of NPCs derived using these protocols is not well defined. Therefore, this study aimed to generate age- and region-specific NPCs from hiPSCs, which mimic in vivo fetal brain (FNPC-B) or spinal cord (FNPC-SC) tissues, in the absence or presence of retinoic acid (RA) and sonic hedgehog (SHH). MATERIALS AND METHODS Ventral, caudal and posterior neural cells were generated from hiPSCs with morphogens (RA and SHH), or dorsal, rostral, and anterior neural cells by the withdrawal of these morphogens from the NPC-media. NPCs generated from hiPSCs were compared to FNPC-B or FNPC-SC using immunocytochemical staining assays and global microarray for the evaluations of general and region-specific neural cells markers of neural induction, differentiation, and maturation. Microarray profiling results were analyzed using quantitative unpaired t-test (P < 0.05). RESULTS Immunocytochemical analyzes showed that generated NPCs expressed general neural cells markers (PAX6 and MUSASHI-2). Furthermore, FNPC-B and anterior NPCs were characterized with marked expression of cortical neural cells marker (SOX1) when compared to FNPC-SC and posterior NPCs. Microarray profiling results showed the up-regulation of brain cells markers (EMX2 and PAX6) in FNPC-B and anterior NPCs. Similarly, spinal cord cells markers (COL5A2, HOXB5, HOXB7, HOXB8, HOXC4, and HOXD4) were up-regulated in FNPC-SC and posterior NPCs. CONCLUSION NPCs that mimic in vivo brain and spinal cord cells can be generated from hiPSCs in the absence or presence of RA and SHH.
Collapse
Affiliation(s)
- Akinlolu Abdulazeez Adelaja
- Department of Anatomy, Faculty of Basic Medical Sciences, University of Ilorin, P. M. B. 1515, Ilorin, Kwara State, Nigeria,Address for correspondence: Akinlolu, Abdulazeez Adelaja, Department of Anatomy, Faculty of Basic Medical Sciences, University of Ilorin, P. M. B. 1515, Ilorin, Kwara State, Nigeria. Phone: 2348062765308. E-mail:
| |
Collapse
|
27
|
Abstract
The ability to use induced pluripotent stem cells (iPSC) to model brain diseases is a powerful tool for unraveling mechanistic alterations in these disorders. Rodent models of brain diseases have spurred understanding of pathology but the concern arises that they may not recapitulate the full spectrum of neuron disruptions associated with human neuropathology. iPSC derived neurons, or other neural cell types, provide the ability to access pathology in cells derived directly from a patient's blood sample or skin biopsy where availability of brain tissue is limiting. Thus, utilization of iPSC to study brain diseases provides an unlimited resource for disease modelling but may also be used for drug screening for effective therapies and may potentially be used to regenerate aged or damaged cells in the future. Many brain diseases across the spectrum of neurodevelopment, neurodegenerative and neuropsychiatric are being approached by iPSC models. The goal of an iPSC based disease model is to identify a cellular phenotype that discriminates the disease-bearing cells from the control cells. In this mini-review, the importance of iPSC cell models validated for pluripotency, germline competency and function assessments is discussed. Selected examples for the variety of brain diseases that are being approached by iPSC technology to discover or establish the molecular basis of the neuropathology are discussed.
Collapse
Affiliation(s)
- Cindy E McKinney
- iPSC Lab/Edward Via College of Osteopathic Medicine and The Gibbs Research Institute, Spartanburg, SC, USA
| |
Collapse
|
28
|
Begum AN, Aguilar JS, Elias L, Hong Y. Silver nanoparticles exhibit coating and dose-dependent neurotoxicity in glutamatergic neurons derived from human embryonic stem cells. Neurotoxicology 2016; 57:45-53. [PMID: 27593553 DOI: 10.1016/j.neuro.2016.08.015] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2016] [Revised: 08/30/2016] [Accepted: 08/31/2016] [Indexed: 10/21/2022]
Abstract
Silver nanoparticles (AgNPs) are used extensively as anti-microbial agents in various products, but little is known about their potential neurotoxic effects. In this study, we used glutamatergic neurons derived from human embryonic stem cells as a cellular model to study 20nm citrate-coated AgNPs (AgSCs) and Polyvinylpyrrolidone-coated AgNPs (AgSPs) induced neurotoxicity. AgSCs significantly damaged neurite outgrowths; increased the production of reactive oxygen species and Ca2+ influxes; reduced the expression of MAP2, PSD95, vGlut1 and NMDA receptor proteins at concentrations as low as 0.1μg/ml. In contrast, AgSPs exhibited neurotoxicity only at higher concentration. Furthermore, our results showed that AgSCs induced glutamate excitotoxicity by the activation of calmodulin and the induction of nitric oxide synthase; increased the phosphorylation of glycogen synthase kinase-3 α/β at Tyr216 and Tau at Ser396 and reduced the expression of Tau46, which are typically observed in Alzheimer's disease. This study indicated that stem cells can provide an excellent platform for studying nanoparticle induced neurotoxicity.
Collapse
Affiliation(s)
- Aynun N Begum
- College of Veterinary Medicine, Western University of Health Sciences, Pomona, CA 91766-1854, USA
| | - Jose S Aguilar
- College of Veterinary Medicine, Western University of Health Sciences, Pomona, CA 91766-1854, USA
| | - Lourdes Elias
- College of Veterinary Medicine, Western University of Health Sciences, Pomona, CA 91766-1854, USA
| | - Yiling Hong
- College of Veterinary Medicine, Western University of Health Sciences, Pomona, CA 91766-1854, USA; Graduate College of Biomedical Sciences, Western University of Health Sciences, Pomona, CA 91766-1854, USA.
| |
Collapse
|
29
|
Li Z, Oganesyan D, Mooney R, Rong X, Christensen MJ, Shahmanyan D, Perrigue PM, Benetatos J, Tsaturyan L, Aramburo S, Annala AJ, Lu Y, Najbauer J, Wu X, Barish ME, Brody DL, Aboody KS, Gutova M. L-MYC Expression Maintains Self-Renewal and Prolongs Multipotency of Primary Human Neural Stem Cells. Stem Cell Reports 2016; 7:483-495. [PMID: 27546534 PMCID: PMC5031988 DOI: 10.1016/j.stemcr.2016.07.013] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2015] [Revised: 07/15/2016] [Accepted: 07/16/2016] [Indexed: 11/25/2022] Open
Abstract
Pre-clinical studies indicate that neural stem cells (NSCs) can limit or reverse CNS damage through direct cell replacement, promotion of regeneration, or delivery of therapeutic agents. Immortalized NSC lines are in growing demand due to the inherent limitations of adult patient-derived NSCs, including availability, expandability, potential for genetic modifications, and costs. Here, we describe the generation and characterization of a new human fetal NSC line, immortalized by transduction with L-MYC (LM-NSC008) that in vitro displays both self-renewal and multipotent differentiation into neurons, oligodendrocytes, and astrocytes. These LM-NSC008 cells were non-tumorigenic in vivo, and migrated to orthotopic glioma xenografts in immunodeficient mice. When administered intranasally, LM-NSC008 distributed specifically to sites of traumatic brain injury (TBI). These data support the therapeutic development of immortalized LM-NSC008 cells for allogeneic use in TBI and other CNS diseases. The generation of a new human fetal L-MYC-immortalized NSC line is described These NSCs display self-renewal and can differentiate into neurons and glia The NSCs can target glioma xenografts and sites of traumatic brain injury in mice This NSC line may become applicable in therapy of various CNS diseases
Collapse
Affiliation(s)
- Zhongqi Li
- Department of Developmental and Stem Cell Biology, City of Hope National Medical Center and Beckman Research Institute of City of Hope, 1500 East Duarte Road, Duarte, CA 91010, USA
| | - Diana Oganesyan
- Department of Developmental and Stem Cell Biology, City of Hope National Medical Center and Beckman Research Institute of City of Hope, 1500 East Duarte Road, Duarte, CA 91010, USA
| | - Rachael Mooney
- Department of Developmental and Stem Cell Biology, City of Hope National Medical Center and Beckman Research Institute of City of Hope, 1500 East Duarte Road, Duarte, CA 91010, USA
| | - Xianfang Rong
- Department of Neurology, Hope Center for Neurological Disorders, Washington University, St. Louis, MO 63110, USA
| | - Matthew J Christensen
- Department of Developmental and Stem Cell Biology, City of Hope National Medical Center and Beckman Research Institute of City of Hope, 1500 East Duarte Road, Duarte, CA 91010, USA
| | - David Shahmanyan
- Department of Developmental and Stem Cell Biology, City of Hope National Medical Center and Beckman Research Institute of City of Hope, 1500 East Duarte Road, Duarte, CA 91010, USA
| | - Patrick M Perrigue
- Department of Epigenetics, Institute of Bioorganic Chemistry of the Polish Academy of Sciences, 61-704 Poznan, Poland
| | - Joseph Benetatos
- Department of Neurology, Hope Center for Neurological Disorders, Washington University, St. Louis, MO 63110, USA
| | - Lusine Tsaturyan
- Department of Developmental and Stem Cell Biology, City of Hope National Medical Center and Beckman Research Institute of City of Hope, 1500 East Duarte Road, Duarte, CA 91010, USA
| | - Soraya Aramburo
- Department of Developmental and Stem Cell Biology, City of Hope National Medical Center and Beckman Research Institute of City of Hope, 1500 East Duarte Road, Duarte, CA 91010, USA
| | - Alexander J Annala
- Department of Developmental and Stem Cell Biology, City of Hope National Medical Center and Beckman Research Institute of City of Hope, 1500 East Duarte Road, Duarte, CA 91010, USA
| | - Yang Lu
- Integrative Genomics Core, City of Hope National Medical Center and Beckman Research Institute of City of Hope, Duarte, CA 91010, USA
| | - Joseph Najbauer
- Department of Immunology and Biotechnology, University of Pécs Medical School, Pécs 7624, Hungary
| | - Xiwei Wu
- Integrative Genomics Core, City of Hope National Medical Center and Beckman Research Institute of City of Hope, Duarte, CA 91010, USA
| | - Michael E Barish
- Department of Developmental and Stem Cell Biology, City of Hope National Medical Center and Beckman Research Institute of City of Hope, 1500 East Duarte Road, Duarte, CA 91010, USA
| | - David L Brody
- Department of Neurology, Hope Center for Neurological Disorders, Washington University, St. Louis, MO 63110, USA
| | - Karen S Aboody
- Department of Developmental and Stem Cell Biology, City of Hope National Medical Center and Beckman Research Institute of City of Hope, 1500 East Duarte Road, Duarte, CA 91010, USA
| | - Margarita Gutova
- Department of Developmental and Stem Cell Biology, City of Hope National Medical Center and Beckman Research Institute of City of Hope, 1500 East Duarte Road, Duarte, CA 91010, USA.
| |
Collapse
|
30
|
Begum AN, Hong Y. A Novel Method for the Generation of Region-Specific Neurons and Neural Networks from Human Pluripotent Stem Cells. JOURNAL OF STEM CELL RESEARCH & THERAPY 2016; 1:1-3. [PMID: 33409004 PMCID: PMC7784482] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Stem cell-based neuronal differentiation has provided a unique opportunity for disease modeling and regenerative medicine. We have reported a novel culture condition and method for generating neuronal progenitors and neural networks from human embryonic and induced pluripotent stem cells without any genetic manipulation. Neurospheres generated under 10% CO2 with Supplemented Knockout Serum Replacement Medium (SKSRM) had doubled the expression of NESTIN, PAX6 and FOXG1 genes compared to the neurospheres generated under 5% CO2. Furthermore, an additional step (AdStep) was introduced to fragment the neurospheres, which increased the expression of neuronal progenitor genes NEUROD1, NEUROG2, TBR1, TBR2, and NOTCH1 and the formation of the neuroepithelial-type cells. With the supplements, neuronal progenitors further differentiated into different layers of cortical, pyramidal, GABAergic, glutamatergic, cholinergic, dopaminergic and purkinje neurons within 27-40 days, which is faster than traditional neurodifferentiation protocols (42-60 days). Furthermore, our in vivo studies indicated that neuronal progenitors derived under our culture conditions with "AdStep" showed significantly increased neurogenesis in Severe Combined Immunodeficiency (SCID) mouse brains. This neurosphere-based neurodifferentiation protocol is a valuable tool for studies neurogenesis, neuronal transplantation and high throughput screening assays.
Collapse
Affiliation(s)
- Aynun N Begum
- College of Veterinary Medicine, Western University of Health Sciences, Pomona, CA, USA
| | - Yiling Hong
- College of Veterinary Medicine, Western University of Health Sciences, Pomona, CA, USA
- Graduate College of Biomedical Sciences, Western University of Health Sciences, Pomona, CA, USA
| |
Collapse
|