1
|
Förster N, Isosaari L, Kulta O, Junnila O, Vuolanto V, Pollari M, Rautajoki KJ, Narkilahti S. Functional 3D Human Neuron-Glioblastoma Model Reveals Cellular Interactions Enabling Drug Safety Assessments. FASEB J 2025; 39:e70567. [PMID: 40277152 PMCID: PMC12023715 DOI: 10.1096/fj.202500291rr] [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: 01/31/2025] [Revised: 04/04/2025] [Accepted: 04/14/2025] [Indexed: 04/26/2025]
Abstract
Glioblastoma (GB) cells actively interact with the central nervous system (CNS) tumor microenvironment (TME). These interactions, particularly with neurons, require a better understanding. 3D tumor models replicating the human TME are needed to unravel pathological processes and to test novel treatments for efficacy and safety. We developed a novel 3D human coculture model for studying neuron-GB interactions. The model revealed both structural and functional interactions between cell types. Paracrine communication in the coculture model favored a tumor-supportive environment. Notably, cell-specific calcium signaling characteristics differed in cocultures compared to monocultures, highlighting the impact of interactions on cellular functionality in TME. The safety of a clinically used treatment, temozolomide, was tested in the 3D coculture model, and it selectively inhibited GB invasion while preserving neurons' morphology and functionality. The established model provides a tool for dissecting the interactions within the TME and testing the efficacy and safety of novel treatments.
Collapse
Affiliation(s)
- Nanna Förster
- NeuroGroup, Faculty of Medicine and Health TechnologyTampere UniversityTampereFinland
| | - Lotta Isosaari
- NeuroGroup, Faculty of Medicine and Health TechnologyTampere UniversityTampereFinland
| | - Oskari Kulta
- NeuroGroup, Faculty of Medicine and Health TechnologyTampere UniversityTampereFinland
| | - Oona Junnila
- NeuroGroup, Faculty of Medicine and Health TechnologyTampere UniversityTampereFinland
| | - Valtteri Vuolanto
- NeuroGroup, Faculty of Medicine and Health TechnologyTampere UniversityTampereFinland
| | - Marjukka Pollari
- Department of Oncology, Tays Cancer CenterTampere University HospitalTampereFinland
| | - Kirsi J. Rautajoki
- Cancer Regulation and Immunology Group, Faculty of Medicine and Health TechnologyTampere UniversityTampereFinland
- Tays Cancer CentreTampere University HospitalTampereFinland
| | - Susanna Narkilahti
- NeuroGroup, Faculty of Medicine and Health TechnologyTampere UniversityTampereFinland
| |
Collapse
|
2
|
Kapucu FE, Tujula I, Kulta O, Sukki L, Ryynänen T, Gram H, Vuolanto V, Vinogradov A, Kreutzer J, Jensen PH, Kallio P, Narkilahti S. Human tripartite cortical network model for temporal assessment of alpha-synuclein aggregation and propagation in Parkinson's Disease. NPJ Parkinsons Dis 2024; 10:138. [PMID: 39069518 DOI: 10.1038/s41531-024-00750-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2023] [Accepted: 07/11/2024] [Indexed: 07/30/2024] Open
Abstract
Previous studies have shown that aggregated alpha-synuclein (α-s) protein, a key pathological marker of Parkinson's disease (PD), can propagate between cells, thus participating in disease progression. This prion-like propagation has been widely studied using in vivo and in vitro models, including rodent and human cell cultures. In this study, our focus was on temporal assessment of functional changes during α-s aggregation and propagation in human induced pluripotent stem cell (hiPSC)-derived neuronal cultures and in engineered networks. Here, we report an engineered circular tripartite human neuronal network model in a microfluidic chip integrated with microelectrode arrays (MEAs) as a platform to study functional markers during α-s aggregation and propagation. We observed progressive aggregation of α-s in conventional neuronal cultures and in the exposed (proximal) compartments of circular tripartite networks following exposure to preformed α-s fibrils (PFF). Furthermore, aggregated forms propagated to distal compartments of the circular tripartite networks through axonal transport. We observed impacts of α-s aggregation on both the structure and function of neuronal cells, such as in presynaptic proteins, mitochondrial motility, calcium oscillations and neuronal activity. The model enabled an assessment of the early, middle, and late phases of α-s aggregation and its propagation during a 13-day follow-up period. While our temporal analysis suggested a complex interplay of structural and functional changes during the in vitro propagation of α-s aggregates, further investigation is required to elucidate the underlying mechanisms. Taken together, this study demonstrates the technical potential of our introduced model for conducting in-depth analyses for revealing such mechanisms.
Collapse
Affiliation(s)
- Fikret Emre Kapucu
- Neuro Group, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland.
| | - Iisa Tujula
- Neuro Group, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Oskari Kulta
- Neuro Group, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Lassi Sukki
- Micro- and Nanosystems Research Group, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Tomi Ryynänen
- Micro- and Nanosystems Research Group, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Hjalte Gram
- Danish Research Institute of Translational Neuroscience - DANDRITE and Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Valtteri Vuolanto
- Neuro Group, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Andrey Vinogradov
- Neuro Group, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Joose Kreutzer
- Micro- and Nanosystems Research Group, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Poul Henning Jensen
- Danish Research Institute of Translational Neuroscience - DANDRITE and Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Pasi Kallio
- Micro- and Nanosystems Research Group, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Susanna Narkilahti
- Neuro Group, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| |
Collapse
|
3
|
Mörö A, Samanta S, Honkamäki L, Rangasami VK, Puistola P, Kauppila M, Narkilahti S, Miettinen S, Oommen O, Skottman H. Hyaluronic acid based next generation bioink for 3D bioprinting of human stem cell derived corneal stromal model with innervation. Biofabrication 2022; 15. [PMID: 36579828 DOI: 10.1088/1758-5090/acab34] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Accepted: 12/13/2022] [Indexed: 12/15/2022]
Abstract
Corneal transplantation remains gold standard for the treatment of severe cornea diseases, however, scarcity of donor cornea is a serious bottleneck. 3D bioprinting holds tremendous potential for cornea tissue engineering (TE). One of the key technological challenges is to design bioink compositions with ideal printability and cytocompatibility. Photo-crosslinking and ionic crosslinking are often used for the stabilization of 3D bioprinted structures, which can possess limitations on biological functionality of the printed cells. Here, we developed a hyaluronic acid-based dopamine containing bioink using hydrazone crosslinking chemistry for the 3D bioprinting of corneal equivalents. First, the shear thinning property, viscosity, and mechanical stability of the bioink were optimized before extrusion-based 3D bioprinting for the shape fidelity and self-healing property characterizations. Subsequently, human adipose stem cells (hASCs) and hASC-derived corneal stromal keratocytes were used for bioprinting corneal stroma structures and their cell viability, proliferation, microstructure and expression of key proteins (lumican, vimentin, connexin 43,α-smooth muscle actin) were evaluated. Moreover, 3D bioprinted stromal structures were implanted intoex vivoporcine cornea to explore tissue integration. Finally, human pluripotent stem cell derived neurons (hPSC-neurons), were 3D bioprinted to the periphery of the corneal structures to analyze innervation. The bioink showed excellent shear thinning property, viscosity, printability, shape fidelity and self-healing properties with high cytocompatibility. Cells in the printed structures displayed good tissue formation and 3D bioprinted cornea structures demonstrated excellentex vivointegration to host tissue as well asin vitroinnervation. The developed bioink and the printed cornea stromal equivalents hold great potential for cornea TE applications.
Collapse
Affiliation(s)
- Anni Mörö
- Eye Regeneration Group, Faculty of Medicine and Health Technology, Tampere University, Tampere 33520, Finland
| | - Sumanta Samanta
- Bioengineering and Nanomedicine Lab, Faculty of Medicine and Health Technology, University, Tampere 33720, Finland
| | - Laura Honkamäki
- Neuro Group, Faculty of Medicine and Health Technology, Tampere University, Tampere 33520, Finland
| | - Vignesh K Rangasami
- Bioengineering and Nanomedicine Lab, Faculty of Medicine and Health Technology, University, Tampere 33720, Finland
| | - Paula Puistola
- Eye Regeneration Group, Faculty of Medicine and Health Technology, Tampere University, Tampere 33520, Finland
| | - Maija Kauppila
- Eye Regeneration Group, Faculty of Medicine and Health Technology, Tampere University, Tampere 33520, Finland
| | - Susanna Narkilahti
- Neuro Group, Faculty of Medicine and Health Technology, Tampere University, Tampere 33520, Finland
| | - Susanna Miettinen
- Adult Stem Cell Group, Faculty of Medicine and Health Technology, Tampere University, Tampere 33520, Finland.,Research, Development and Innovation Centre, Tampere University Hospital, Tampere 33520, Finland
| | - Oommen Oommen
- Bioengineering and Nanomedicine Lab, Faculty of Medicine and Health Technology, University, Tampere 33720, Finland
| | - Heli Skottman
- Eye Regeneration Group, Faculty of Medicine and Health Technology, Tampere University, Tampere 33520, Finland
| |
Collapse
|
4
|
Lotila J, Hyvärinen T, Skottman H, Airas L, Narkilahti S, Hagman S. Establishment of a human induced pluripotent stem cell line (TAUi008-A) derived from a multiple sclerosis patient. Stem Cell Res 2022; 63:102865. [PMID: 35843021 DOI: 10.1016/j.scr.2022.102865] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Accepted: 07/08/2022] [Indexed: 10/17/2022] Open
Abstract
Multiple sclerosis (MS) is a complex autoimmune disease of the central nervous system where the main pathogenetic events include demyelination and axonal degeneration. Here, we generated a human induced pluripotent stem cell (hiPSC) line from peripheral blood mononuclear cells of an MS patient utilizing Sendai virus reprogramming. The produced hiPSC line expressed pluripotency markers, differentiated into three germ layers, showed a normal karyotype and was free of virus vectors, transgenes and mycoplasma. Established hiPSCs are a valuable source for studies of MS disease modeling and drug discovery.
Collapse
Affiliation(s)
- Johanna Lotila
- Neuroimmunology Research Group, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland; NeuroGroup, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Tanja Hyvärinen
- Neuroimmunology Research Group, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Heli Skottman
- Eye Group, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Laura Airas
- Clinical Neurosciences, University of Turku and Neurocenter, Turku University Hospital, Turku, Finland
| | - Susanna Narkilahti
- NeuroGroup, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Sanna Hagman
- Neuroimmunology Research Group, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland.
| |
Collapse
|
5
|
A kainic acid-induced seizure model in human pluripotent stem cell-derived cortical neurons for studying the role of IL-6 in the functional activity. Stem Cell Res 2022; 60:102665. [DOI: 10.1016/j.scr.2022.102665] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Revised: 01/09/2022] [Accepted: 01/11/2022] [Indexed: 11/20/2022] Open
|
6
|
Functional Characterization of Human Pluripotent Stem Cell-Derived Models of the Brain with Microelectrode Arrays. Cells 2021; 11:cells11010106. [PMID: 35011667 PMCID: PMC8750870 DOI: 10.3390/cells11010106] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Revised: 12/22/2021] [Accepted: 12/24/2021] [Indexed: 12/26/2022] Open
Abstract
Human pluripotent stem cell (hPSC)-derived neuron cultures have emerged as models of electrical activity in the human brain. Microelectrode arrays (MEAs) measure changes in the extracellular electric potential of cell cultures or tissues and enable the recording of neuronal network activity. MEAs have been applied to both human subjects and hPSC-derived brain models. Here, we review the literature on the functional characterization of hPSC-derived two- and three-dimensional brain models with MEAs and examine their network function in physiological and pathological contexts. We also summarize MEA results from the human brain and compare them to the literature on MEA recordings of hPSC-derived brain models. MEA recordings have shown network activity in two-dimensional hPSC-derived brain models that is comparable to the human brain and revealed pathology-associated changes in disease models. Three-dimensional hPSC-derived models such as brain organoids possess a more relevant microenvironment, tissue architecture and potential for modeling the network activity with more complexity than two-dimensional models. hPSC-derived brain models recapitulate many aspects of network function in the human brain and provide valid disease models, but certain advancements in differentiation methods, bioengineering and available MEA technology are needed for these approaches to reach their full potential.
Collapse
|
7
|
Honkamäki L, Joki T, Grigoryev NA, Levon K, Ylä-Outinen L, Narkilahti S. Novel method to produce a layered 3D scaffold for human pluripotent stem cell-derived neuronal cells. J Neurosci Methods 2020; 350:109043. [PMID: 33345946 DOI: 10.1016/j.jneumeth.2020.109043] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Revised: 12/10/2020] [Accepted: 12/11/2020] [Indexed: 01/13/2023]
Abstract
BACKGROUND Three-dimensional (3D) in vitro models have been developed into more in vivo resembling structures. In particular, there is a need for human-based models for neuronal tissue engineering (TE). To produce such a model with organized microenvironment for cells in central nervous system (CNS), a 3D layered scaffold composed of hydrogel and cell guiding fibers has been proposed. NEW METHOD Here, we describe a novel method for producing a layered 3D scaffold consisting of electrospun poly (L,D-lactide) fibers embedded into collagen 1 hydrogel to achieve better resemblance of cells' natural microenvironment for human pluripotent stem cell (hPSC)-derived neurons. The scaffold was constructed via a single layer-by-layer process using an electrospinning technique with a unique collector design. RESULTS The method enabled the production of layered 3D cell-containing scaffold in a single process. HPSC-derived neurons were found in all layers of the scaffold and exhibited a typical neuronal phenotype. The guiding fiber layers supported the directed cell growth and extension of the neurites inside the scaffold without additional functionalization. COMPARISON WITH EXISTING METHODS Previous methods have required several process steps to construct 3D layer-by-layer scaffolds. CONCLUSIONS We introduced a method to produce layered 3D scaffolds to mimic the cell guiding cues in CNS by alternating the soft hydrogel matrix and fibrous guidance cues. The produced scaffold successfully enabled the long-term culture of hPSC-derived neuronal cells. This layered 3D scaffold is a useful model for in vitro and in vivo neuronal TE applications.
Collapse
Affiliation(s)
- Laura Honkamäki
- NeuroGroup, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland.
| | - Tiina Joki
- NeuroGroup, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Nikita A Grigoryev
- Department of Chemical and Biomolecular Engineering, NYU Tandon School of Engineering, Brooklyn, NY, USA
| | - Kalle Levon
- Department of Chemical and Biomolecular Engineering, NYU Tandon School of Engineering, Brooklyn, NY, USA
| | - Laura Ylä-Outinen
- NeuroGroup, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Susanna Narkilahti
- NeuroGroup, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| |
Collapse
|
8
|
Pelkonen A, Mzezewa R, Sukki L, Ryynänen T, Kreutzer J, Hyvärinen T, Vinogradov A, Aarnos L, Lekkala J, Kallio P, Narkilahti S. A modular brain-on-a-chip for modelling epileptic seizures with functionally connected human neuronal networks. Biosens Bioelectron 2020; 168:112553. [DOI: 10.1016/j.bios.2020.112553] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Revised: 08/01/2020] [Accepted: 08/23/2020] [Indexed: 12/22/2022]
|
9
|
In Vitro Oxygen-Glucose Deprivation-Induced Stroke Models with Human Neuroblastoma Cell- and Induced Pluripotent Stem Cell-Derived Neurons. Stem Cells Int 2020; 2020:8841026. [PMID: 33178286 PMCID: PMC7647751 DOI: 10.1155/2020/8841026] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Revised: 10/07/2020] [Accepted: 10/07/2020] [Indexed: 12/20/2022] Open
Abstract
Stroke is a devastating neurological disorder and one of the leading causes of mortality and disability. To understand the cellular and molecular mechanisms of stroke and to develop novel therapeutic approaches, two different in vitro human cell-based stroke models were established using oxygen-glucose deprivation (OGD) conditions. In addition, the effect of adipose stem cells (ASCs) on OGD-induced injury was studied. In the present study, SH-SY5Y human neuroblastoma cells and human induced pluripotent stem cells (hiPSCs) were differentiated into neurons, cultured under OGD conditions (1% O2) for 24 h, and subjected to a reperfusion period for 24 or 72 h. After OGD, ASCs were cocultured with neurons on inserts for 24 or 72 h to study the neuroprotective potential of ASCs. The effect of OGD and ASC coculture on the viability, apoptosis, and proliferation of and axonal damage to neuronal cells was studied. The results showed that OGD conditions induced cytotoxicity and apoptosis of SH-SY5Y- and hiPSC-derived neurons, although more severe damage was detected in SH-SY5Y-derived neurons than in hiPSC-derived neurons. Coculture with ASCs was protective for neurons, as the number of dead ASC-cocultured neurons was lower than that of control cells, and coculture increased the proliferation of both cell types. To conclude, we developed in vitro human cell-based stroke models in SH-SY5Y- and hiPSC-derived neurons. This was the first time hiPSCs were used to model stroke in vitro. Since OGD had different effects on the studied cell types, this study highlights the importance of using several cell types in in vitro studies to confirm the outcomes of the study. Here, ASCs exerted a neuroprotective effect by increasing the proliferation and decreasing the death of SH-SY5Y- and hiPSC-derived neurons after OGD.
Collapse
|
10
|
Välimäki H, Hyvärinen T, Leivo J, Iftikhar H, Pekkanen-Mattila M, Rajan DK, Verho J, Kreutzer J, Ryynänen T, Pirhonen J, Aalto-Setälä K, Kallio P, Narkilahti S, Lekkala J. Covalent immobilization of luminescent oxygen indicators reduces cytotoxicity. Biomed Microdevices 2020; 22:41. [PMID: 32494857 PMCID: PMC7270993 DOI: 10.1007/s10544-020-00495-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Luminescence-based oxygen sensing is a widely used tool in cell culture applications. In a typical configuration, the luminescent oxygen indicators are embedded in a solid, oxygen-permeable matrix in contact with the culture medium. However, in sensitive cell cultures even minimal leaching of the potentially cytotoxic indicators can become an issue. One way to prevent the leaching is to immobilize the indicators covalently into the supporting matrix. In this paper, we report on a method where platinum(II)-5,10,15,20-tetrakis-(2,3,4,5,6-pentafluorphenyl)-porphyrin (PtTFPP) oxygen indicators are covalently immobilized into a polymer matrix consisting of polystyrene and poly(pentafluorostyrene). We study how the covalent immobilization influences the sensing material’s cytotoxicity to human induced pluripotent stem cell-derived (hiPSC-derived) neurons and cardiomyocytes (CMs) through 7–13 days culturing experiments and various viability analyses. Furthermore, we study the effect of the covalent immobilization on the indicator leaching and the oxygen sensing properties of the material. In addition, we demonstrate the use of the covalently linked oxygen sensing material in real time oxygen tension monitoring in functional hypoxia studies of the hiPSC-derived CMs. The results show that the covalently immobilized indicators substantially reduce indicator leaching and the cytotoxicity of the oxygen sensing material, while the influence on the oxygen sensing properties remains small or nonexistent.
Collapse
Affiliation(s)
- Hannu Välimäki
- Faculty of Medicine and Health Technology, Tampere University, Korkeakoulunkatu 3, 33720, Tampere, Finland.
| | - Tanja Hyvärinen
- Faculty of Medicine and Health Technology, Tampere University, Arvo Ylpön katu 34, 33520, Tampere, Finland
| | - Joni Leivo
- Faculty of Medicine and Health Technology, Tampere University, Korkeakoulunkatu 3, 33720, Tampere, Finland
| | - Haider Iftikhar
- Faculty of Medicine and Health Technology, Tampere University, Korkeakoulunkatu 3, 33720, Tampere, Finland
| | - Mari Pekkanen-Mattila
- Faculty of Medicine and Health Technology, Tampere University, Arvo Ylpön katu 34, 33520, Tampere, Finland
| | | | - Jarmo Verho
- Faculty of Medicine and Health Technology, Tampere University, Korkeakoulunkatu 3, 33720, Tampere, Finland
| | - Joose Kreutzer
- Faculty of Medicine and Health Technology, Tampere University, Korkeakoulunkatu 3, 33720, Tampere, Finland
| | - Tomi Ryynänen
- Faculty of Medicine and Health Technology, Tampere University, Korkeakoulunkatu 3, 33720, Tampere, Finland
| | - Jonatan Pirhonen
- Faculty of Medicine and Health Technology, Tampere University, Arvo Ylpön katu 34, 33520, Tampere, Finland
| | - Katriina Aalto-Setälä
- Faculty of Medicine and Health Technology, Tampere University, Arvo Ylpön katu 34, 33520, Tampere, Finland
| | - Pasi Kallio
- Faculty of Medicine and Health Technology, Tampere University, Korkeakoulunkatu 3, 33720, Tampere, Finland
| | - Susanna Narkilahti
- Faculty of Medicine and Health Technology, Tampere University, Arvo Ylpön katu 34, 33520, Tampere, Finland
| | - Jukka Lekkala
- Faculty of Medicine and Health Technology, Tampere University, Korkeakoulunkatu 3, 33720, Tampere, Finland
| |
Collapse
|
11
|
Hyvärinen T, Hyysalo A, Kapucu FE, Aarnos L, Vinogradov A, Eglen SJ, Ylä-Outinen L, Narkilahti S. Functional characterization of human pluripotent stem cell-derived cortical networks differentiated on laminin-521 substrate: comparison to rat cortical cultures. Sci Rep 2019; 9:17125. [PMID: 31748598 PMCID: PMC6868015 DOI: 10.1038/s41598-019-53647-8] [Citation(s) in RCA: 67] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Accepted: 11/01/2019] [Indexed: 12/15/2022] Open
Abstract
Human pluripotent stem cell (hPSC)-derived neurons provide exciting opportunities for in vitro modeling of neurological diseases and for advancing drug development and neurotoxicological studies. However, generating electrophysiologically mature neuronal networks from hPSCs has been challenging. Here, we report the differentiation of functionally active hPSC-derived cortical networks on defined laminin-521 substrate. We apply microelectrode array (MEA) measurements to assess network events and compare the activity development of hPSC-derived networks to that of widely used rat embryonic cortical cultures. In both of these networks, activity developed through a similar sequence of stages and time frames; however, the hPSC-derived networks showed unique patterns of bursting activity. The hPSC-derived networks developed synchronous activity, which involved glutamatergic and GABAergic inputs, recapitulating the classical cortical activity also observed in rodent counterparts. Principal component analysis (PCA) based on spike rates, network synchronization and burst features revealed the segregation of hPSC-derived and rat network recordings into different clusters, reflecting the species-specific and maturation state differences between the two networks. Overall, hPSC-derived neural cultures produced with a defined protocol generate cortical type network activity, which validates their applicability as a human-specific model for pharmacological studies and modeling network dysfunctions.
Collapse
Affiliation(s)
- Tanja Hyvärinen
- Faculty of Medicine and Health Technology and BioMediTech, Tampere University, Tampere, Finland
| | - Anu Hyysalo
- Faculty of Medicine and Health Technology and BioMediTech, Tampere University, Tampere, Finland
- Institute of Biotechnology, HiLIFE, University of Helsinki, Helsinki, Finland
| | - Fikret Emre Kapucu
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
- Danish Research Institute of Translational Neuroscience - DANDRITE, Aarhus University, Aarhus, Denmark
| | - Laura Aarnos
- Faculty of Medicine and Health Technology and BioMediTech, Tampere University, Tampere, Finland
| | - Andrey Vinogradov
- Faculty of Medicine and Health Technology and BioMediTech, Tampere University, Tampere, Finland
| | - Stephen J Eglen
- Department of Applied Mathematics and Theoretical Physics, University of Cambridge, Cambridge, United Kingdom
| | - Laura Ylä-Outinen
- Faculty of Medicine and Health Technology and BioMediTech, Tampere University, Tampere, Finland
| | - Susanna Narkilahti
- Faculty of Medicine and Health Technology and BioMediTech, Tampere University, Tampere, Finland.
| |
Collapse
|
12
|
Hyvärinen T, Hagman S, Ristola M, Sukki L, Veijula K, Kreutzer J, Kallio P, Narkilahti S. Co-stimulation with IL-1β and TNF-α induces an inflammatory reactive astrocyte phenotype with neurosupportive characteristics in a human pluripotent stem cell model system. Sci Rep 2019; 9:16944. [PMID: 31729450 PMCID: PMC6858358 DOI: 10.1038/s41598-019-53414-9] [Citation(s) in RCA: 108] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Accepted: 10/31/2019] [Indexed: 12/14/2022] Open
Abstract
Astrocyte reactivation has been discovered to be an important contributor to several neurological diseases. In vitro models involving human astrocytes have the potential to reveal disease-specific mechanisms of these cells and to advance research on neuropathological conditions. Here, we induced a reactive phenotype in human induced pluripotent stem cell (hiPSC)-derived astrocytes and studied the inflammatory natures and effects of these cells on human neurons. Astrocytes responded to interleukin-1β (IL-1β) and tumor necrosis factor-α (TNF-α) treatment with a typical transition to polygonal morphology and a shift to an inflammatory phenotype characterized by altered gene and protein expression profiles. Astrocyte-secreted factors did not exert neurotoxic effects, whereas they transiently promoted the functional activity of neurons. Importantly, we engineered a novel microfluidic platform designed for investigating interactions between neuronal axons and reactive astrocytes that also enables the implementation of a controlled inflammatory environment. In this platform, selective stimulation of astrocytes resulted in an inflammatory niche that sustained axonal growth, further suggesting that treatment induces a reactive astrocyte phenotype with neurosupportive characteristics. Our findings show that hiPSC-derived astrocytes are suitable for modeling astrogliosis, and the developed in vitro platform provides promising novel tools for studying neuron-astrocyte crosstalk and human brain disease in a dish.
Collapse
Affiliation(s)
- Tanja Hyvärinen
- NeuroGroup, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Sanna Hagman
- NeuroGroup, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Mervi Ristola
- NeuroGroup, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Lassi Sukki
- Micro and Nanosystems Research Group, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Katariina Veijula
- NeuroGroup, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Joose Kreutzer
- Micro and Nanosystems Research Group, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Pasi Kallio
- Micro and Nanosystems Research Group, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Susanna Narkilahti
- NeuroGroup, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland.
| |
Collapse
|
13
|
Ylä‐Outinen L, Harju V, Joki T, Koivisto JT, Karvinen J, Kellomäki M, Narkilahti S. Screening of Hydrogels for Human Pluripotent Stem Cell–Derived Neural Cells: Hyaluronan‐Polyvinyl Alcohol‐Collagen‐Based Interpenetrating Polymer Network Provides an Improved Hydrogel Scaffold. Macromol Biosci 2019; 19:e1900096. [DOI: 10.1002/mabi.201900096] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Revised: 05/24/2019] [Indexed: 12/11/2022]
Affiliation(s)
- Laura Ylä‐Outinen
- NeuroGroup, Faculty of Medicine and Health TechnologyTampere University Kalevantie 4 33014 Tampere Finland
| | - Venla Harju
- NeuroGroup, Faculty of Medicine and Health TechnologyTampere University Kalevantie 4 33014 Tampere Finland
| | - Tiina Joki
- NeuroGroup, Faculty of Medicine and Health TechnologyTampere University Kalevantie 4 33014 Tampere Finland
| | - Janne T. Koivisto
- Biomaterials and Tissue Engineering GroupFaculty of Medicine and Health TechnologyTampere University Kalevantie 4 33014 Tampere Finland
| | - Jennika Karvinen
- Biomaterials and Tissue Engineering GroupFaculty of Medicine and Health TechnologyTampere University Kalevantie 4 33014 Tampere Finland
| | - Minna Kellomäki
- Biomaterials and Tissue Engineering GroupFaculty of Medicine and Health TechnologyTampere University Kalevantie 4 33014 Tampere Finland
| | - Susanna Narkilahti
- NeuroGroup, Faculty of Medicine and Health TechnologyTampere University Kalevantie 4 33014 Tampere Finland
| |
Collapse
|
14
|
Ryynänen T, Pelkonen A, Grigoras K, Ylivaara OME, Hyvärinen T, Ahopelto J, Prunnila M, Narkilahti S, Lekkala J. Microelectrode Array With Transparent ALD TiN Electrodes. Front Neurosci 2019; 13:226. [PMID: 30967754 PMCID: PMC6438859 DOI: 10.3389/fnins.2019.00226] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Accepted: 02/26/2019] [Indexed: 11/16/2022] Open
Abstract
Low noise platinum black or sputtered titanium nitride (TiN) microelectrodes are typically used for recording electrical activity of neuronal or cardiac cell cultures. Opaque electrodes and tracks, however, hinder the visibility of the cells when imaged with inverted microscope, which is the standard method of imaging cells plated on microelectrode array (MEA). Even though transparent indium tin oxide (ITO) electrodes exist, they cannot compete in impedance and noise performance with above-mentioned opaque counterparts. In this work, we propose atomic layer deposition (ALD) as the method to deposit TiN electrodes and tracks which are thin enough (25–65 nm) to be transparent (transmission ∼18–45%), but still benefit from the columnar structure of TiN, which is the key element to decrease noise and impedance of the electrodes. For ALD TiN electrodes (diameter 30 μm) impedances from 510 to 590 kΩ were measured at 1 kHz, which is less than the impedance of bare ITO electrodes. Human induced pluripotent stem cell (hiPSC)-derived cortical neurons were cultured on the ALD TiN MEAs for 14 days without observing any biocompatibility issues, and spontaneous electrical activity of the neurons was recorded successfully. The results show that transparent ALD TiN film is a suitable electrode material for producing functional MEAs.
Collapse
Affiliation(s)
- Tomi Ryynänen
- Micro- and Nanosystems Research Group, BioMediTech Institute and Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Anssi Pelkonen
- NeuroGroup, BioMediTech Institute and Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | | | | | - Tanja Hyvärinen
- NeuroGroup, BioMediTech Institute and Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Jouni Ahopelto
- VTT Technical Research Centre of Finland Ltd., Espoo, Finland
| | - Mika Prunnila
- VTT Technical Research Centre of Finland Ltd., Espoo, Finland
| | - Susanna Narkilahti
- NeuroGroup, BioMediTech Institute and Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Jukka Lekkala
- Micro- and Nanosystems Research Group, BioMediTech Institute and Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| |
Collapse
|
15
|
Ylä-Outinen L, Tanskanen JMA, Kapucu FE, Hyysalo A, Hyttinen JAK, Narkilahti S. Advances in Human Stem Cell-Derived Neuronal Cell Culturing and Analysis. ADVANCES IN NEUROBIOLOGY 2019; 22:299-329. [DOI: 10.1007/978-3-030-11135-9_13] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
16
|
Ryynänen T, Toivanen M, Salminen T, Ylä-Outinen L, Narkilahti S, Lekkala J. Ion Beam Assisted E-Beam Deposited TiN Microelectrodes-Applied to Neuronal Cell Culture Medium Evaluation. Front Neurosci 2018; 12:882. [PMID: 30568570 PMCID: PMC6290344 DOI: 10.3389/fnins.2018.00882] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2017] [Accepted: 11/12/2018] [Indexed: 12/15/2022] Open
Abstract
Microelectrode material and cell culture medium have significant roles in the signal-to-noise ratio and cell well-being in in vitro electrophysiological studies. Here, we report an ion beam assisted e-beam deposition (IBAD) based process as an alternative titanium nitride (TiN) deposition method for sputtering in the fabrication of state-of-the-art TiN microelectrode arrays (MEAs). The effects of evaporation and nitrogen flow rates were evaluated while developing the IBAD TiN deposition process. Moreover, the produced IBAD TiN microelectrodes were characterized by impedance, charge transfer capacity (CTC) and noise measurements for electrical properties, AFM and SEM for topological imaging, and EDS for material composition. The impedance (at 1 kHz) of brand new 30 μm IBAD TiN microelectrodes was found to be double but still below 100 kΩ compared with commercial reference MEAs with sputtered TiN microelectrodes of the same size. On the contrary, the noise level of IBAD TiN MEAs was lower compared with that of commercial sputtered TiN MEAs in equal conditions. In CTC IBAD TiN electrodes (3.3 mC/cm2) also outperformed the sputtered counterparts (2.0 mC/cm2). To verify the suitability of IBAD TiN microelectrodes for cell measurements, human pluripotent stem cell (hPSC)-derived neuronal networks were cultured on IBAD TiN MEAs and commercial sputtered TiN MEAs in two different media: neural differentiation medium (NDM) and BrainPhys (BPH). The effect of cell culture media to hPSC derived neuronal networks was evaluated to gain more stable and more active networks. Higher spontaneous activity levels were measured from the neuronal networks cultured in BPH compared with those in NDM in both MEA types. However, BPH caused more problems in cell survival in long-term cultures by inducing neuronal network retraction and clump formation after 1–2 weeks. In addition, BPH was found to corrode the Si3N4 insulator layer more than NDM medium. The developed IBAD TiN process gives MEA manufacturers more choices to choose which method to use to deposit TiN electrodes and the medium evaluation results remind that not only electrode material but also insulator layer and cell culturing medium have crucial role in successful long term MEA measurements.
Collapse
Affiliation(s)
- Tomi Ryynänen
- BioMediTech Institute and Faculty of Biomedical Sciences and Engineering, Tampere University of Technology, Tampere, Finland
| | - Maria Toivanen
- NeuroGroup, BioMediTech Institute and Faculty of Medicine and Life Sciences, University of Tampere, Tampere, Finland
| | - Turkka Salminen
- Laboratory of Photonics, Tampere University of Technology, Tampere, Finland
| | - Laura Ylä-Outinen
- NeuroGroup, BioMediTech Institute and Faculty of Medicine and Life Sciences, University of Tampere, Tampere, Finland
| | - Susanna Narkilahti
- NeuroGroup, BioMediTech Institute and Faculty of Medicine and Life Sciences, University of Tampere, Tampere, Finland
| | - Jukka Lekkala
- BioMediTech Institute and Faculty of Biomedical Sciences and Engineering, Tampere University of Technology, Tampere, Finland
| |
Collapse
|
17
|
Liu Y, Antonic A, Yang X, Korte N, Lim K, Michalska AE, Dottori M, Howells DW. Derivation of phenotypically diverse neural culture from hESC by combining adherent and dissociation methods. J Neurosci Methods 2018; 308:286-293. [PMID: 30003885 DOI: 10.1016/j.jneumeth.2018.07.005] [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: 01/13/2018] [Revised: 07/05/2018] [Accepted: 07/05/2018] [Indexed: 11/18/2022]
Abstract
BACKGROUND Differentiation of human embryonic stem cells (hESCs) into distinct neural lineages has been widely studied. However, preparation of mixed yet neurochemically mature populations, for the study of neurological diseases involving mixed cell types has received less attention. NEW METHOD We combined two commonly used differentiation methods to provide robust and reproducible cultures in which a mixture of primarily GABAergic and Glutamatergic neurons was obtained. Detailed characterisation by immunocytochemistry (ICC) and quantitative real-time PCR (qPCR) assessed the neurochemical phenotype, and the maturation state of these neurons. RESULTS We found that once neurospheres (NSs) had attached to the culture plates, proliferation of neural stem cell was suppressed. Neuronal differentiation and synaptic development then occurred after 21 days in vitro (DIV). By 49DIV, there were large numbers of neurochemically and structurally mature neurons. The qPCR studies indicated that expression of GABAergic genes increased the most (93.3-fold increase), followed by glutamatergic (51-fold increase), along with smaller changes in expression of cholinergic (3-fold increase) and dopaminergic genes (6-fold increase), as well as a small change in glial cell marker expression (5-fold increase). COMPARISON WITH EXISTING METHOD (S) Existing methods isolate hESC-derived neural progenitors for onward differentiation to mature neurons using either migration or dissociative paradigms. These give poor survival or yield. By combining these approaches, we obtain high yields of morphologically and neurochemically mature neurons. These can be maintained in culture for extended periods. CONCLUSION Our method provides a novel, effective and robust neural culture system with structurally and neurochemically mature cell populations and neural networks, suitable for studying a range of neurological diseases from a human perspective.
Collapse
Affiliation(s)
- Ye Liu
- Florey Institute of Neuroscience and Mental Health, 30 Royal Parade, The University of Melbourne, Victoria, 3010, Australia; Department of Neurology, Fudan University, Huashan Hospital, Shanghai, 200040, China
| | - Ana Antonic
- Department of Neuroscience, Central Clinical School, Monash University, The Alfred Centre, VIC, 3004, Australia
| | - Xuan Yang
- Institute for Geriatrics and Rehabilitation, Beijing Geriatric Hospital, Beijing, 100095, China
| | - Nils Korte
- Department of Neuroscience, Physiology and Pharmacology, University College London, Gower Street, London, WC1E6BT, UK
| | - Katherine Lim
- Stem Cell Core Facility, Stem Cells Australia, The University of Melbourne, Victoria, 3010, Australia
| | - Anna E Michalska
- Stem Cell Core Facility, Stem Cells Australia, The University of Melbourne, Victoria, 3010, Australia
| | - Mirella Dottori
- Illawarra Health and Medical Research Institute Centre for Molecular and Medical Bioscience Building 32, University of Wollongong, NSW, 2522 Australia
| | - David W Howells
- School of Medicine, University of Tasmania, Hobart, Tasmania, 7001, Australia.
| |
Collapse
|
18
|
Effects of inflammatory cytokines IFN-γ, TNF-α and IL-6 on the viability and functionality of human pluripotent stem cell-derived neural cells. J Neuroimmunol 2018; 331:36-45. [PMID: 30195439 DOI: 10.1016/j.jneuroim.2018.07.010] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2017] [Revised: 07/13/2018] [Accepted: 07/23/2018] [Indexed: 02/07/2023]
Abstract
Multiple Sclerosis (MS) is an inflammatory neurodegenerative disease, where neural progenitor cell (NPC) transplantation has been suggested as a potential neuroprotective therapeutic strategy. Since the effect of inflammation on NPCs is poorly known, their effect on the survival and functionality of human NPCs were studied. Treatment with interleukin (IL)-6, tumor necrosis factor (TNF)-α and interferon (IFN)-γ did not induced cytotoxicity, but IFN-γ treatment showed decreased proliferation and neuronal migration. By contrast, increased proliferation and inhibition of electrical activity were detected after TNF-α treatment. Treatments induced secretion of inflammatory factors. Inflammatory cytokines appear to modulate proliferation as well as the cellular and functional properties of human NPCs.
Collapse
|
19
|
Mäkinen MEL, Ylä-Outinen L, Narkilahti S. GABA and Gap Junctions in the Development of Synchronized Activity in Human Pluripotent Stem Cell-Derived Neural Networks. Front Cell Neurosci 2018; 12:56. [PMID: 29559893 PMCID: PMC5845705 DOI: 10.3389/fncel.2018.00056] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2017] [Accepted: 02/16/2018] [Indexed: 01/03/2023] Open
Abstract
The electrical activity of the brain arises from single neurons communicating with each other. However, how single neurons interact during early development to give rise to neural network activity remains poorly understood. We studied the emergence of synchronous neural activity in human pluripotent stem cell (hPSC)-derived neural networks simultaneously on a single-neuron level and network level. The contribution of gamma-aminobutyric acid (GABA) and gap junctions to the development of synchronous activity in hPSC-derived neural networks was studied with GABA agonist and antagonist and by blocking gap junctional communication, respectively. We characterized the dynamics of the network-wide synchrony in hPSC-derived neural networks with high spatial resolution (calcium imaging) and temporal resolution microelectrode array (MEA). We found that the emergence of synchrony correlates with a decrease in very strong GABA excitation. However, the synchronous network was found to consist of a heterogeneous mixture of synchronously active cells with variable responses to GABA, GABA agonists and gap junction blockers. Furthermore, we show how single-cell distributions give rise to the network effect of GABA, GABA agonists and gap junction blockers. Finally, based on our observations, we suggest that the earliest form of synchronous neuronal activity depends on gap junctions and a decrease in GABA induced depolarization but not on GABAA mediated signaling.
Collapse
Affiliation(s)
- Meeri Eeva-Liisa Mäkinen
- NeuroGroup Laboratory, BioMediTech Institute and Faculty of Medicine and Life Sciences, University of Tampere, Tampere, Finland
| | - Laura Ylä-Outinen
- NeuroGroup Laboratory, BioMediTech Institute and Faculty of Medicine and Life Sciences, University of Tampere, Tampere, Finland
| | - Susanna Narkilahti
- NeuroGroup Laboratory, BioMediTech Institute and Faculty of Medicine and Life Sciences, University of Tampere, Tampere, Finland
| |
Collapse
|
20
|
Williams M, Prem S, Zhou X, Matteson P, Yeung PL, Lu CW, Pang Z, Brzustowicz L, Millonig JH, Dicicco-Bloom E. Rapid Detection of Neurodevelopmental Phenotypes in Human Neural Precursor Cells (NPCs). J Vis Exp 2018. [PMID: 29553565 DOI: 10.3791/56628] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Human brain development proceeds through a series of precisely orchestrated processes, with earlier stages distinguished by proliferation, migration, and neurite outgrowth; and later stages characterized by axon/dendrite outgrowth and synapse formation. In neurodevelopmental disorders, often one or more of these processes are disrupted, leading to abnormalities in brain formation and function. With the advent of human induced pluripotent stem cell (hiPSC) technology, researchers now have an abundant supply of human cells that can be differentiated into virtually any cell type, including neurons. These cells can be used to study both normal brain development and disease pathogenesis. A number of protocols using hiPSCs to model neuropsychiatric disease use terminally differentiated neurons or use 3D culture systems termed organoids. While these methods have proven invaluable in studying human disease pathogenesis, there are some drawbacks. Differentiation of hiPSCs into neurons and generation of organoids are lengthy and costly processes that can impact the number of experiments and variables that can be assessed. In addition, while post-mitotic neurons and organoids allow the study of disease-related processes, including dendrite outgrowth and synaptogenesis, they preclude the study of earlier processes like proliferation and migration. In neurodevelopmental disorders, such as autism, abundant genetic and post-mortem evidence indicates defects in early developmental processes. Neural precursor cells (NPCs), a highly proliferative cell population, may be a suitable model in which to ask questions about ontogenetic processes and disease initiation. We now extend methodologies learned from studying development in mouse and rat cortical cultures to human NPCs. The use of NPCs allows us to investigate disease-related phenotypes and define how different variables (e.g., growth factors, drugs) impact developmental processes including proliferation, migration, and differentiation in only a few days. Ultimately, this toolset can be used in a reproducible and high-throughput manner to identify disease-specific mechanisms and phenotypes in neurodevelopmental disorders.
Collapse
Affiliation(s)
- Madeline Williams
- Department of Neuroscience and Cell Biology, Rutgers Robert Wood Johnson Medical School
| | - Smrithi Prem
- Department of Neuroscience and Cell Biology, Rutgers Robert Wood Johnson Medical School
| | - Xiaofeng Zhou
- Department of Neuroscience and Cell Biology, Rutgers Robert Wood Johnson Medical School
| | - Paul Matteson
- Center for Advanced Biotechnology and Medicine, Department of Neuroscience and Cell Biology, Rutgers Robert Wood Johnson Medical School
| | - Percy Luk Yeung
- The Child Health Institute of NJ, Department of Obstetrics, Gynecology, and Reproductive Services, Rutgers Robert Wood Johnson Medical School
| | - Chi-Wei Lu
- The Child Health Institute of NJ, Department of Obstetrics, Gynecology, and Reproductive Services, Rutgers Robert Wood Johnson Medical School
| | - Zhiping Pang
- The Child Health Institute of NJ, Department of Neuroscience and Cell Biology, Rutgers Robert Wood Johnson Medical School
| | | | - James H Millonig
- Center for Advanced Biotechnology and Medicine, Department of Neuroscience and Cell Biology, Rutgers Robert Wood Johnson Medical School
| | - Emanuel Dicicco-Bloom
- Department of Neuroscience and Cell Biology, Rutgers Robert Wood Johnson Medical School;
| |
Collapse
|
21
|
Karvinen J, Joki T, Ylä-Outinen L, Koivisto JT, Narkilahti S, Kellomäki M. Soft hydrazone crosslinked hyaluronan- and alginate-based hydrogels as 3D supportive matrices for human pluripotent stem cell-derived neuronal cells. REACT FUNCT POLYM 2018. [DOI: 10.1016/j.reactfunctpolym.2017.12.019] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
|
22
|
Effect of prolonged differentiation on functional maturation of human pluripotent stem cell-derived neuronal cultures. Stem Cell Res 2018; 27:151-161. [PMID: 29414606 DOI: 10.1016/j.scr.2018.01.018] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2017] [Revised: 01/09/2018] [Accepted: 01/17/2018] [Indexed: 01/15/2023] Open
Abstract
Long-term neural differentiation of human pluripotent stem cells (hPSCs) is associated with enhanced neuronal maturation, which is a necessity for creation of representative in vitro models. It also induces neurogenic-to-gliogenic fate switch, increasing proportion of endogenous astrocytes formed from the common neural progenitors. However, the significance of prolonged differentiation on the neural cell type composition and functional development of hPSC-derived neuronal cells has not been well characterized. Here, we studied two hPSC lines, both of which initially showed good neuronal differentiation capacity. However, the propensity for endogenous astrogenesis and maturation state after extended differentiation varied. Live cell calcium imaging revealed that prolonged differentiation facilitated maturation of GABAergic signaling. According to extracellular recordings with microelectrode array (MEA), neuronal activity was limited to fewer areas of the culture, which expressed more frequent burst activity. Efficient maturation after prolonged differentiation also promoted organization of spontaneous activity by burst compaction. These results suggest that although prolonged neural differentiation can be challenging, it has beneficial effect on functional maturation, which can also improve transition to different neural in vitro models and applications.
Collapse
|
23
|
Toivanen M, Pelkonen A, Mäkinen M, Ylä-Outinen L, Sukki L, Kallio P, Ristola M, Narkilahti S. Optimised PDMS Tunnel Devices on MEAs Increase the Probability of Detecting Electrical Activity from Human Stem Cell-Derived Neuronal Networks. Front Neurosci 2017; 11:606. [PMID: 29163011 PMCID: PMC5671636 DOI: 10.3389/fnins.2017.00606] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2017] [Accepted: 10/17/2017] [Indexed: 12/22/2022] Open
Abstract
Measurement of the activity of human pluripotent stem cell (hPSC)-derived neuronal networks with microelectrode arrays (MEAs) plays an important role in functional in vitro brain modelling and in neurotoxicological screening. The previously reported hPSC-derived neuronal networks do not, however, exhibit repeatable, stable functional network characteristics similar to rodent cortical cultures, making the interpretation of results difficult. In earlier studies, microtunnels have been used both to control and guide cell growth and amplify the axonal signals of rodent neurons. The aim of the current study was to develop tunnel devices that would facilitate signalling and/or signal detection in entire hPSC-derived neuronal networks containing not only axons, but also somata and dendrites. Therefore, MEA-compatible polydimethylsiloxane (PDMS) tunnel devices with 8 different dimensions were created. The hPSC-derived neurons were cultured in the tunnel devices on MEAs, and the spontaneous electrical activity of the networks was measured for 5 weeks. Although the tunnel devices improved the signal-to-noise ratio only by 1.3-fold at best, they significantly increased the percentage of electrodes detecting neuronal activity (52–100%) compared with the controls (27%). Significantly higher spike and burst counts were also obtained using the tunnel devices. Neuronal networks inside the tunnels were amenable to pharmacological manipulation. The results suggest that tunnel devices encompassing the entire neuronal network can increase the measured spontaneous activity in hPSC-derived neuronal networks on MEAs. Therefore, they can increase the efficiency of functional studies of hPSC-derived networks on MEAs.
Collapse
Affiliation(s)
- Maria Toivanen
- NeuroGroup, BioMediTech Institute and Faculty of Medicine and Biosciences, University of Tampere, Tampere, Finland
| | - Anssi Pelkonen
- NeuroGroup, BioMediTech Institute and Faculty of Medicine and Biosciences, University of Tampere, Tampere, Finland
| | - Meeri Mäkinen
- NeuroGroup, BioMediTech Institute and Faculty of Medicine and Biosciences, University of Tampere, Tampere, Finland
| | - Laura Ylä-Outinen
- NeuroGroup, BioMediTech Institute and Faculty of Medicine and Biosciences, University of Tampere, Tampere, Finland
| | - Lassi Sukki
- Micro and Nanosystems Research Group, BioMediTech Institute and Faculty of Biomedical Sciences and Engineering, Tampere University of Technology, Tampere, Finland
| | - Pasi Kallio
- Micro and Nanosystems Research Group, BioMediTech Institute and Faculty of Biomedical Sciences and Engineering, Tampere University of Technology, Tampere, Finland
| | - Mervi Ristola
- NeuroGroup, BioMediTech Institute and Faculty of Medicine and Biosciences, University of Tampere, Tampere, Finland
| | - Susanna Narkilahti
- NeuroGroup, BioMediTech Institute and Faculty of Medicine and Biosciences, University of Tampere, Tampere, Finland
| |
Collapse
|
24
|
Hyysalo A, Ristola M, Mäkinen MEL, Häyrynen S, Nykter M, Narkilahti S. Laminin α5 substrates promote survival, network formation and functional development of human pluripotent stem cell-derived neurons in vitro. Stem Cell Res 2017; 24:118-127. [DOI: 10.1016/j.scr.2017.09.002] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2016] [Revised: 08/28/2017] [Accepted: 09/07/2017] [Indexed: 01/24/2023] Open
|
25
|
Turunen S, Joki T, Hiltunen ML, Ihalainen TO, Narkilahti S, Kellomäki M. Direct Laser Writing of Tubular Microtowers for 3D Culture of Human Pluripotent Stem Cell-Derived Neuronal Cells. ACS APPLIED MATERIALS & INTERFACES 2017; 9:25717-25730. [PMID: 28697300 DOI: 10.1021/acsami.7b05536] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
As the complex structure of nervous tissue cannot be mimicked in two-dimensional (2D) cultures, the development of three-dimensional (3D) neuronal cell culture platforms is a topical issue in the field of neuroscience and neural tissue engineering. Computer-assisted laser-based fabrication techniques such as direct laser writing by two-photon polymerization (2PP-DLW) offer a versatile tool to fabricate 3D cell culture platforms with highly ordered geometries in the size scale of natural 3D cell environments. In this study, we present the design and 2PP-DLW fabrication process of a novel 3D neuronal cell culture platform based on tubular microtowers. The platform facilitates efficient long-term 3D culturing of human neuronal cells and supports neurite orientation and 3D network formation. Microtower designs both with or without intraluminal guidance cues and/or openings in the tower wall are designed and successfully fabricated from Ormocomp. Three of the microtower designs are chosen for the final culture platform: a design with openings in the wall and intralumial guidance cues (webs and pillars), a design with openings but without intraluminal structures, and a plain cylinder design. The proposed culture platform offers a promising concept for future 3D cultures in the field of neuroscience.
Collapse
Affiliation(s)
- Sanna Turunen
- Biomaterials and Tissue Engineering Group, BioMediTech and Faculty of Biomedical Sciences and Engineering, Tampere University of Technology , Korkeakoulunkatu 3, 33720 Tampere, Finland
| | - Tiina Joki
- NeuroGroup, BioMediTech and Faculty of Medicine and Life Sciences, University of Tampere , Lääkärinkatu 1, 33520 Tampere, Finland
| | - Maiju L Hiltunen
- Biomaterials and Tissue Engineering Group, BioMediTech and Faculty of Biomedical Sciences and Engineering, Tampere University of Technology , Korkeakoulunkatu 3, 33720 Tampere, Finland
| | - Teemu O Ihalainen
- NeuroGroup, BioMediTech and Faculty of Medicine and Life Sciences, University of Tampere , Lääkärinkatu 1, 33520 Tampere, Finland
| | - Susanna Narkilahti
- NeuroGroup, BioMediTech and Faculty of Medicine and Life Sciences, University of Tampere , Lääkärinkatu 1, 33520 Tampere, Finland
| | - Minna Kellomäki
- Biomaterials and Tissue Engineering Group, BioMediTech and Faculty of Biomedical Sciences and Engineering, Tampere University of Technology , Korkeakoulunkatu 3, 33720 Tampere, Finland
- BioMediTech and Faculty of Medicine and Life Sciences, University of Tampere , Lääkärinkatu 1, 33520 Tampere, Finland
| |
Collapse
|
26
|
Koivisto JT, Joki T, Parraga JE, Pääkkönen R, Ylä-Outinen L, Salonen L, Jönkkäri I, Peltola M, Ihalainen TO, Narkilahti S, Kellomäki M. Bioamine-crosslinked gellan gum hydrogel for neural tissue engineering. Biomed Mater 2017; 12:025014. [DOI: 10.1088/1748-605x/aa62b0] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
|
27
|
Hyysalo A, Ristola M, Joki T, Honkanen M, Vippola M, Narkilahti S. Aligned Poly(ε-caprolactone) Nanofibers Guide the Orientation and Migration of Human Pluripotent Stem Cell-Derived Neurons, Astrocytes, and Oligodendrocyte Precursor Cells In Vitro. Macromol Biosci 2017; 17. [DOI: 10.1002/mabi.201600517] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2016] [Revised: 02/06/2017] [Indexed: 12/31/2022]
Affiliation(s)
- Anu Hyysalo
- NeuroGroup; BioMediTech and Faculty of Medicine and Life Sciences; University of Tampere; Lääkärinkatu 1 FI-33520 Tampere Finland
| | - Mervi Ristola
- NeuroGroup; BioMediTech and Faculty of Medicine and Life Sciences; University of Tampere; Lääkärinkatu 1 FI-33520 Tampere Finland
| | - Tiina Joki
- NeuroGroup; BioMediTech and Faculty of Medicine and Life Sciences; University of Tampere; Lääkärinkatu 1 FI-33520 Tampere Finland
| | - Mari Honkanen
- Department of Materials Science; Tampere University of Technology; Korkeakoulunkatu 6 FI-33720 Tampere Finland
| | - Minnamari Vippola
- Department of Materials Science; Tampere University of Technology; Korkeakoulunkatu 6 FI-33720 Tampere Finland
| | - Susanna Narkilahti
- NeuroGroup; BioMediTech and Faculty of Medicine and Life Sciences; University of Tampere; Lääkärinkatu 1 FI-33520 Tampere Finland
| |
Collapse
|
28
|
Cell culture chamber with gas supply for prolonged recording of human neuronal cells on microelectrode array. J Neurosci Methods 2017; 280:27-35. [DOI: 10.1016/j.jneumeth.2017.01.019] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2016] [Revised: 01/26/2017] [Accepted: 01/31/2017] [Indexed: 01/02/2023]
|
29
|
Lukovic D, Diez Lloret A, Stojkovic P, Rodríguez-Martínez D, Perez Arago MA, Rodriguez-Jimenez FJ, González-Rodríguez P, López-Barneo J, Sykova E, Jendelova P, Kostic J, Moreno-Manzano V, Stojkovic M, Bhattacharya SS, Erceg S. Highly Efficient Neural Conversion of Human Pluripotent Stem Cells in Adherent and Animal-Free Conditions. Stem Cells Transl Med 2017; 6:1217-1226. [PMID: 28213969 PMCID: PMC5442830 DOI: 10.1002/sctm.16-0371] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2016] [Revised: 10/20/2016] [Accepted: 11/16/2016] [Indexed: 12/23/2022] Open
Abstract
Neural differentiation of human embryonic stem cells (hESCs) and induced pluripotent stem cells (hiPSCs) can produce a valuable and robust source of human neural cell subtypes, holding great promise for the study of neurogenesis and development, and for treating neurological diseases. However, current hESCs and hiPSCs neural differentiation protocols require either animal factors or embryoid body formation, which decreases efficiency and yield, and strongly limits medical applications. Here we develop a simple, animal-free protocol for neural conversion of both hESCs and hiPSCs in adherent culture conditions. A simple medium formula including insulin induces the direct conversion of >98% of hESCs and hiPSCs into expandable, transplantable, and functional neural progenitors with neural rosette characteristics. Further differentiation of neural progenitors into dopaminergic and spinal motoneurons as well as astrocytes and oligodendrocytes indicates that these neural progenitors retain responsiveness to instructive cues revealing the robust applicability of the protocol in the treatment of different neurodegenerative diseases. The fact that this protocol includes animal-free medium and human extracellular matrix components avoiding embryoid bodies makes this protocol suitable for the use in clinic. Stem Cells Translational Medicine 2017;6:1217-1226.
Collapse
Affiliation(s)
- Dunja Lukovic
- Stem Cells Therapies in Neurodegenerative Diseases Lab.,National Stem Cell Bank-Valencia Node, Biomolecular and Bioinformatics Resources Platform PRB2,ISCIII
| | - Andrea Diez Lloret
- CABIMER (Centro Andaluz de Biología Molecular y Medicina Regenerativa), Avda. Americo Vespucio s/n, Parque Científico y Tecnológico Cartuja, Sevilla, Spain
| | | | - Daniel Rodríguez-Martínez
- CABIMER (Centro Andaluz de Biología Molecular y Medicina Regenerativa), Avda. Americo Vespucio s/n, Parque Científico y Tecnológico Cartuja, Sevilla, Spain
| | | | | | - Patricia González-Rodríguez
- Instituto de Biomedicina de Sevilla (IBiS) and Departamento de Fisiología Médica y Biofísica, Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Seville, Spain
| | - José López-Barneo
- Instituto de Biomedicina de Sevilla (IBiS) and Departamento de Fisiología Médica y Biofísica, Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Seville, Spain
| | - Eva Sykova
- Department of Neuroscience, Institute of Experimental Medicine, Academy of Science of the Czech Republic, Prague, Czech Republic
| | - Pavla Jendelova
- Department of Neuroscience, Institute of Experimental Medicine, Academy of Science of the Czech Republic, Prague, Czech Republic
| | - Jelena Kostic
- Stem Cells Therapies in Neurodegenerative Diseases Lab
| | | | - Miodrag Stojkovic
- Spebo Medical, Leskovac, Serbia.,Faculty of Medical Sciences, Human Genetics Department, University of Kragujevac, Serbia
| | - Shomi S Bhattacharya
- CABIMER (Centro Andaluz de Biología Molecular y Medicina Regenerativa), Avda. Americo Vespucio s/n, Parque Científico y Tecnológico Cartuja, Sevilla, Spain
| | - Slaven Erceg
- Stem Cells Therapies in Neurodegenerative Diseases Lab.,National Stem Cell Bank-Valencia Node, Biomolecular and Bioinformatics Resources Platform PRB2,ISCIII.,Department of Neuroscience, Institute of Experimental Medicine, Academy of Science of the Czech Republic, Prague, Czech Republic
| |
Collapse
|
30
|
Lenk K, Priwitzer B, Ylä-Outinen L, Tietz LHB, Narkilahti S, Hyttinen JAK. Simulation of developing human neuronal cell networks. Biomed Eng Online 2016; 15:105. [PMID: 27576323 PMCID: PMC5006268 DOI: 10.1186/s12938-016-0226-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2015] [Accepted: 08/18/2016] [Indexed: 12/15/2022] Open
Abstract
Background Microelectrode array (MEA) is a widely used technique to study for example the functional properties of neuronal networks derived from human embryonic stem cells (hESC-NN). With hESC-NN, we can investigate the earliest developmental stages of neuronal network formation in the human brain. Methods In this paper, we propose an in silico model of maturating hESC-NNs based on a phenomenological model called INEX. We focus on simulations of the development of bursts in hESC-NNs, which are the main feature of neuronal activation patterns. The model was developed with data from developing hESC-NN recordings on MEAs which showed increase in the neuronal activity during the investigated six measurement time points in the experimental and simulated data. Results Our simulations suggest that the maturation process of hESC-NN, resulting in the formation of bursts, can be explained by the development of synapses. Moreover, spike and burst rate both decreased at the last measurement time point suggesting a pruning of synapses as the weak ones are removed. Conclusions To conclude, our model reflects the assumption that the interaction between excitatory and inhibitory neurons during the maturation of a neuronal network and the spontaneous emergence of bursts are due to increased connectivity caused by the forming of new synapses.
Collapse
Affiliation(s)
- Kerstin Lenk
- Department of Electronics and Communications Engineering, Tampere University of Technology, BioMediTech, PL100, Tampere, Finland.
| | - Barbara Priwitzer
- Faculty of Engineering and Computer Science, Brandenburg University of Technology Cottbus-Senftenberg, Platz der Deutschen Einheit 1, 03046, Cottbus, Germany
| | - Laura Ylä-Outinen
- NeuroGroup, Institute of Biomedical Technology, University of Tampere, BioMediTech, PL100, Tampere, Finland
| | - Lukas H B Tietz
- Department of Electronics and Communications Engineering, Tampere University of Technology, BioMediTech, PL100, Tampere, Finland
| | - Susanna Narkilahti
- NeuroGroup, Institute of Biomedical Technology, University of Tampere, BioMediTech, PL100, Tampere, Finland
| | - Jari A K Hyttinen
- Department of Electronics and Communications Engineering, Tampere University of Technology, BioMediTech, PL100, Tampere, Finland
| |
Collapse
|
31
|
Ensuring the Quality of Stem Cell-Derived In Vitro Models for Toxicity Testing. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2016; 856:259-297. [DOI: 10.1007/978-3-319-33826-2_11] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
32
|
Kapucu FE, Mäkinen MEL, Tanskanen JMA, Ylä-Outinen L, Narkilahti S, Hyttinen JAK. Joint analysis of extracellular spike waveforms and neuronal network bursts. J Neurosci Methods 2015; 259:143-155. [PMID: 26675487 DOI: 10.1016/j.jneumeth.2015.11.022] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2015] [Revised: 11/23/2015] [Accepted: 11/24/2015] [Indexed: 01/13/2023]
Abstract
BACKGROUND Neuronal networks are routinely assessed based on extracellular electrophysiological microelectrode array (MEA) measurements by spike sorting, and spike and burst statistics. We propose to jointly analyze sorted spikes and detected bursts, and hypothesize that the obtained spike type compositions of the bursts can provide new information on the functional networks. NEW METHOD Spikes are detected and sorted to obtain spike types and bursts are detected. In the proposed joint analysis, each burst spike is associated with a spike type, and the spike type compositions of the bursts are assessed. RESULTS The proposed method was tested with simulations and MEA measurements of in vitro human stem cell derived neuronal networks under different pharmacological treatments. The results show that the treatments altered the spike type compositions of the bursts. For example, 6-cyano-7-nitroquinoxaline-2,3-dione almost completely abolished two types of spikes which had composed the bursts in the baseline, while bursts of spikes of two other types appeared more frequently. This phenomenon was not observable by spike sorting or burst analysis alone, but was revealed by the proposed joint analysis. COMPARISON WITH EXISTING METHODS The existing methods do not provide the information obtainable with the proposed method: for the first time, the spike type compositions of bursts are analyzed. CONCLUSIONS We showed that the proposed method provides useful and novel information, including the possible changes in the spike type compositions of the bursts due to external factors. Our method can be employed on any data exhibiting sortable action potential waveforms and detectable bursts.
Collapse
Affiliation(s)
- Fikret Emre Kapucu
- Tampere University of Technology, Department of Electronics and Communications Engineering, Computational Biophysics and Imaging Group, BioMediTech, Biokatu 6, FI-33520 Tampere, Finland.
| | - Meeri E-L Mäkinen
- University of Tampere, NeuroGroup, BioMediTech, Biokatu 12, FI-33014 Tampere, Finland.
| | - Jarno M A Tanskanen
- Tampere University of Technology, Department of Electronics and Communications Engineering, Computational Biophysics and Imaging Group, BioMediTech, Biokatu 6, FI-33520 Tampere, Finland.
| | - Laura Ylä-Outinen
- University of Tampere, NeuroGroup, BioMediTech, Biokatu 12, FI-33014 Tampere, Finland.
| | - Susanna Narkilahti
- University of Tampere, NeuroGroup, BioMediTech, Biokatu 12, FI-33014 Tampere, Finland.
| | - Jari A K Hyttinen
- Tampere University of Technology, Department of Electronics and Communications Engineering, Computational Biophysics and Imaging Group, BioMediTech, Biokatu 6, FI-33520 Tampere, Finland.
| |
Collapse
|
33
|
Wu S, Johansson J, Damdimopoulou P, Shahsavani M, Falk A, Hovatta O, Rising A. Spider silk for xeno-free long-term self-renewal and differentiation of human pluripotent stem cells. Biomaterials 2014; 35:8496-502. [PMID: 25043502 DOI: 10.1016/j.biomaterials.2014.06.039] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2014] [Accepted: 06/20/2014] [Indexed: 11/29/2022]
Abstract
Human pluripotent stem cells (hPSCs) can undergo unlimited self-renewal and have the capacity to differentiate into all somatic cell types, and are therefore an ideal source for the generation of cells and tissues for research and therapy. To realize this potential, defined cell culture systems that allow expansion of hPSCs and subsequent controlled differentiation, ideally in an implantable three-dimensional (3D) matrix, are required. Here we mimic spider silk - Nature's high performance material - for the design of chemically defined 2D and 3D matrices for cell culture. The silk matrices do not only allow xeno-free long-term expansion of hPSCs but also differentiation in both 2D and 3D. These results show that biomimetic spider silk matrices enable hPSC culture in a manner that can be applied for experimental and clinical purposes.
Collapse
Affiliation(s)
- Siqin Wu
- Department of Neurobiology, Care Sciences and Society (NVS), Karolinska Institutet, Novum 5th floor, 141 86 Stockholm, Sweden
| | - Jan Johansson
- Department of Neurobiology, Care Sciences and Society (NVS), Karolinska Institutet, Novum 5th floor, 141 86 Stockholm, Sweden; Department of Anatomy, Physiology and Biochemistry, Swedish University of Agricultural Sciences, The Biomedical Centre, Box 575, 751 23 Uppsala, Sweden; Institute of Mathematics and Natural Sciences, Tallinn University, Narva mnt 25, 101 20 Tallinn, Estonia
| | - Pauliina Damdimopoulou
- Department of Clinical Sciences, Intervention and Technology, Division of Obstetrics and Gynecology, Karolinska Institutet and Karolinska University Hospital, Huddinge, 141 86 Stockholm, Sweden
| | - Mansoureh Shahsavani
- Department of Neuroscience, Karolinska Institutet, Retzius v. 8, 171 77 Stockholm, Sweden
| | - Anna Falk
- Department of Neuroscience, Karolinska Institutet, Retzius v. 8, 171 77 Stockholm, Sweden
| | - Outi Hovatta
- Department of Clinical Sciences, Intervention and Technology, Division of Obstetrics and Gynecology, Karolinska Institutet and Karolinska University Hospital, Huddinge, 141 86 Stockholm, Sweden
| | - Anna Rising
- Department of Neurobiology, Care Sciences and Society (NVS), Karolinska Institutet, Novum 5th floor, 141 86 Stockholm, Sweden; Department of Anatomy, Physiology and Biochemistry, Swedish University of Agricultural Sciences, The Biomedical Centre, Box 575, 751 23 Uppsala, Sweden.
| |
Collapse
|
34
|
Translation: screening for novel therapeutics with disease-relevant cell types derived from human stem cell models. Biol Psychiatry 2014; 75:952-60. [PMID: 23876186 PMCID: PMC3815991 DOI: 10.1016/j.biopsych.2013.05.028] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2013] [Revised: 05/02/2013] [Accepted: 05/29/2013] [Indexed: 12/23/2022]
Abstract
The advent of somatic cell reprogramming technologies-which enables the generation of patient-specific, induced pluripotent stem cell and other trans-differentiated human neuronal cell models-provides new means of gaining insight into the molecular mechanisms and neural substrates of psychiatric disorders. By allowing a more precise understanding of genotype-phenotype relationship in disease-relevant human cell types, the use of reprogramming technologies in tandem with emerging genome engineering approaches provides a previously "missing link" between basic research and translational efforts. In this review, we summarize advances in applying human pluripotent stem cell and reprogramming technologies to generate specific neural subtypes with a focus on the use of these in vitro systems for the discovery of small molecule-probes and novel therapeutics. Examples are given where human cell models of psychiatric disorders have begun to reveal new mechanistic insight into pathophysiology and simultaneously have provided the foundation for developing disease-relevant, phenotypic assays suitable for both functional genomic and chemical screens. A number of areas for future research are discussed, including the need to develop robust methodology for the reproducible, large-scale production of disease-relevant neural cell types in formats compatible with high-throughput screening modalities, including high-content imaging, multidimensional, signature-based screening, and in vitro network with multielectrode arrays. Limitations, including the challenges in recapitulating neurocircuits and non-cell autonomous phenotypes are discussed. Although these technologies are still in active development, we conclude that, as our understanding of how to efficiently generate and probe the plasticity of patient-specific stem models improves, their utility is likely to advance rapidly.
Collapse
|
35
|
Sterthaus O, Feutz AC, Zhang H, Pletscher F, Bruder E, Miny P, Lezzi G, De Geyter M, De Geyter C. Gene expression profiles of similarly derived human embryonic stem cell lines correlate with their distinct propensity to exit stemness and their different differentiation behavior in culture. Cell Reprogram 2014; 16:185-95. [PMID: 24811852 DOI: 10.1089/cell.2013.0089] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Four normal-karyotype human embryonic stem cell (hESC) lines were generated using the same protocol and maintained under identical conditions. Despite these precautions, gene expression patterns were found to be dissimilar among the four lines. The observed differences were typical of each cell line, correlated with their distinct propensity to exit stemness, created heterogeneity among the cells during cell line maintenance, and correlated with their altered capacity as a source of differentiated cells. The capacity of some cell lines to give rise to more, and more mature, neurons within comparable time frames of directed differentiation reflected the distinct proportions of cells already predifferentiated at the onset. These findings demonstrate that the subsequent stages of neural differentiation were altered both in a quantitative and timely fashion. As a consequence, cell lines with apparent better and quicker ability to produce neurons were actually the less capable of reproducing proper differentiation. Previous data suggested that cell lines able to generate more neurons faster would be more suitable to clinical application. Our analysis of the differentiation process strongly suggests the opposite. The spontaneous tendency to predifferentiate of any particular hESC line should be known because it clearly impacts further experimental results.
Collapse
Affiliation(s)
- Oliver Sterthaus
- 1 Clinic of Gynecological Endocrinology and Reproductive Medicine, University of Basel , CH-4031, Basel, Switzerland
| | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Ljung K, Simonson OE, Felldin U, Wärdell E, Ibarra C, Antonsson L, Kumagai-Braesch M, Hovatta O, Lampela R, Grinnemo KH, Corbascio M. Costimulation blockade induces foxp3(+) regulatory T cells to human embryonic stem cells. Biores Open Access 2013; 2:455-8. [PMID: 24380056 PMCID: PMC3869426 DOI: 10.1089/biores.2013.0039] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
Transplantation of human embryonic stem cells (hESCs), like other allogeneic cellular transplants, require immunomodulation or immunosuppression in order to be maintained in the recipient. Costimulation blockade applied at the time of transplantation inhibits costimulatory signals in the immunological synapse leading to a state of anergy in the donor reactive T-cell population and a state of immunological tolerance in the host. In models of solid organ transplantation, tolerance is maintained by the infiltration of Foxp3+ regulatory T cells into the graft. In order to study if regulatory T cells could be generated to hESC transplants, costimulation blockade (CTLA4Ig, anti-CD40L, anti-LFA-1) was administered for the first week after transplantation of two different hESC lines implanted under the kidney capsule of wild-type mice. hESC transplants were maintained indefinitely, and when harvested at long-term follow-up, Foxp3+ T-cells were found surrounding the graft, implying the maintenance of tolerance through the induction of regulatory T cells. These results imply that costimulation blockade could be a useful treatment strategy for the induction of tolerance to hESC transplants and may down-modulate immune responses locally around the graft.
Collapse
Affiliation(s)
- Karin Ljung
- Department of Emergency Medicine (Huddinge), Karolinska University Hospital , Stockholm, Sweden
| | - Oscar E Simonson
- Department of Cardiothoracic Surgery & Anesthesiology (Solna), Karolinska University Hospital , Stockholm, Sweden
| | - Ulrika Felldin
- Department of Molecular Medicine & Surgery, Karolinska Institute , Stockholm, Sweden
| | - Ewa Wärdell
- Department of Medicine, Karolinska Institute , Stockholm, Sweden
| | - Cristian Ibarra
- Department of Medical Biochemistry & Biophysics, Karolinska Institute , Stockholm, Sweden
| | - Liselotte Antonsson
- Department of Clinical Science, Intervention, & Technology, Karolinska Institute , Stockholm, Sweden
| | - Makiko Kumagai-Braesch
- Department of Clinical Science, Intervention, & Technology, Karolinska Institute , Stockholm, Sweden
| | - Outi Hovatta
- Department of Clinical Science, Intervention, & Technology, Karolinska Institute , Stockholm, Sweden
| | - Riina Lampela
- Department of Clinical Science, Intervention, & Technology, Karolinska Institute , Stockholm, Sweden
| | - Karl-Henrik Grinnemo
- Department of Cardiothoracic Surgery & Anesthesiology (Solna), Karolinska University Hospital , Stockholm, Sweden
| | - Matthias Corbascio
- Department of Cardiothoracic Surgery & Anesthesiology (Solna), Karolinska University Hospital , Stockholm, Sweden
| |
Collapse
|
37
|
Kolaja K. Stem cells and stem cell-derived tissues and their use in safety assessment. J Biol Chem 2013; 289:4555-61. [PMID: 24362027 DOI: 10.1074/jbc.r113.481028] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Toxicology has long relied on animal models in a tedious approach to understanding risk of exposure to an uncharacterized molecule. Stem cell-derived tissues can be made in high purity, quality, and quantity to enable a new approach to this problem. Currently, stem cell-derived tissues are primarily "generic" genetic backgrounds; the future will see the integration of various genetic backgrounds and complex three-dimensional models to create truly unique in vitro organoids. This minireview focuses on the state of the art of a number of stem cell-derived tissues and details their application in toxicology.
Collapse
Affiliation(s)
- Kyle Kolaja
- From Cellular Dynamics International, Montclair, New Jersey 07042
| |
Collapse
|
38
|
Puttonen KA, Ruponen M, Kauppinen R, Wojciechowski S, Hovatta O, Koistinaho J. Improved Method of Producing Human Neural Progenitor Cells of High Purity and in Large Quantities from Pluripotent Stem Cells for Transplantation Studies. Cell Transplant 2013; 22:1753-66. [DOI: 10.3727/096368912x658764] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
Transplantation of human neural progenitor cells (hNPCs) is a promising therapeutic approach for various diseases of the central nervous system (CNS). Reliable testing of hNPC transplantation in animal models of neurological diseases requires that these cells can be produced in sufficient amounts, show consistent homogeneity as a neural cell population, and be reliably labeled for in vivo tracking. In addition, the cells should be characterized as being at the optimal state of differentiation favoring successful engraftment. Here, we show that high numbers of purified hNPCs can be produced from human embryonic stem cells (hESCs) by manually selecting specifically sized and shaped spheres followed by fluorescence-activated cell sorting based on the relative cell size. In addition, we report that labeling of hNPCs with ultra-small superparamagnetic iron oxide (USPIO) particles does not affect the cellular morphology or growth. More importantly, we show that the transduction with lentiviral vector encoding green fluorescent protein (GFP) decreases the neurality of the cell population. We conclude that our cost-effective protocol of generating hNPCs is widely applicable for preclinical studies on CNS disorders. This improved method of producing large quantities of high-purity hNPCs maybe useful also when generating hNPCs from human induced pluripotent stem (hiPS) cell lines. However, caution should be used when lenti-GFP transduction is applied for hNPC labeling.
Collapse
Affiliation(s)
- Katja A. Puttonen
- Department of Neurobiology, A. I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Marika Ruponen
- Department of Neurobiology, A. I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
- School of Pharmacy, University of Eastern Finland, Kuopio, Finland
| | - Riitta Kauppinen
- Department of Neurobiology, A. I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Sara Wojciechowski
- Department of Neurobiology, A. I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Outi Hovatta
- Department of Biotechnology and Molecular Medicine, A. I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
- Department of Clinical Science, Intervention and Technology, Karolinska Institute, Stockholm, Sweden
| | - Jari Koistinaho
- Department of Neurobiology, A. I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
- Department of Oncology, Kuopio University Hospital, Kuopio, Finland
| |
Collapse
|
39
|
Maria S, Helle B, Tristan L, Gaynor S, Arnar A, Michele M, Teresia O, Oliver C, Roger S, Penelope H, Ole I. Improved cell therapy protocols for Parkinson's disease based on differentiation efficiency and safety of hESC-, hiPSC-, and non-human primate iPSC-derived dopaminergic neurons. Stem Cells 2013; 31:1548-62. [PMID: 23666606 PMCID: PMC3775937 DOI: 10.1002/stem.1415] [Citation(s) in RCA: 171] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2013] [Accepted: 04/01/2013] [Indexed: 12/22/2022]
Abstract
The main motor symptoms of Parkinson's disease are due to the loss of dopaminergic (DA) neurons in the ventral midbrain (VM). For the future treatment of Parkinson's disease with cell transplantation it is important to develop efficient differentiation methods for production of human iPSCs and hESCs-derived midbrain-type DA neurons. Here we describe an efficient differentiation and sorting strategy for DA neurons from both human ES/iPS cells and non-human primate iPSCs. The use of non-human primate iPSCs for neuronal differentiation and autologous transplantation is important for preclinical evaluation of safety and efficacy of stem cell-derived DA neurons. The aim of this study was to improve the safety of human- and non-human primate iPSC (PiPSC)-derived DA neurons. According to our results, NCAM(+) /CD29(low) sorting enriched VM DA neurons from pluripotent stem cell-derived neural cell populations. NCAM(+) /CD29(low) DA neurons were positive for FOXA2/TH and EN1/TH and this cell population had increased expression levels of FOXA2, LMX1A, TH, GIRK2, PITX3, EN1, NURR1 mRNA compared to unsorted neural cell populations. PiPSC-derived NCAM(+) /CD29(low) DA neurons were able to restore motor function of 6-hydroxydopamine (6-OHDA) lesioned rats 16 weeks after transplantation. The transplanted sorted cells also integrated in the rodent brain tissue, with robust TH+/hNCAM+ neuritic innervation of the host striatum. One year after autologous transplantation, the primate iPSC-derived neural cells survived in the striatum of one primate without any immunosuppression. These neural cell grafts contained FOXA2/TH-positive neurons in the graft site. This is an important proof of concept for the feasibility and safety of iPSC-derived cell transplantation therapies in the future.
Collapse
Affiliation(s)
- Sundberg Maria
- Neuroregeneration Laboratories, Harvard Medical School/McLean Hospital, Belmont, MA, 02478
| | - Bogetofte Helle
- Neuroregeneration Laboratories, Harvard Medical School/McLean Hospital, Belmont, MA, 02478
| | - Lawson Tristan
- Neuroregeneration Laboratories, Harvard Medical School/McLean Hospital, Belmont, MA, 02478
- New England Primate Research Center, Harvard Medical School, Southborough, MA 01772
| | - Smith Gaynor
- Neuroregeneration Laboratories, Harvard Medical School/McLean Hospital, Belmont, MA, 02478
| | - Astradsson Arnar
- Neuroregeneration Laboratories, Harvard Medical School/McLean Hospital, Belmont, MA, 02478
| | - Moore Michele
- Neuroregeneration Laboratories, Harvard Medical School/McLean Hospital, Belmont, MA, 02478
- New England Primate Research Center, Harvard Medical School, Southborough, MA 01772
| | - Osborn Teresia
- Neuroregeneration Laboratories, Harvard Medical School/McLean Hospital, Belmont, MA, 02478
| | - Cooper Oliver
- Neuroregeneration Laboratories, Harvard Medical School/McLean Hospital, Belmont, MA, 02478
| | - Spealman Roger
- New England Primate Research Center, Harvard Medical School, Southborough, MA 01772
| | - Hallett Penelope
- Neuroregeneration Laboratories, Harvard Medical School/McLean Hospital, Belmont, MA, 02478
| | - Isacson Ole
- Neuroregeneration Laboratories, Harvard Medical School/McLean Hospital, Belmont, MA, 02478
| |
Collapse
|
40
|
Kiiski H, Aänismaa R, Tenhunen J, Hagman S, Ylä-Outinen L, Aho A, Yli-Hankala A, Bendel S, Skottman H, Narkilahti S. Healthy human CSF promotes glial differentiation of hESC-derived neural cells while retaining spontaneous activity in existing neuronal networks. Biol Open 2013; 2:605-12. [PMID: 23789111 PMCID: PMC3683163 DOI: 10.1242/bio.20134648] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2013] [Accepted: 04/12/2013] [Indexed: 12/19/2022] Open
Abstract
The possibilities of human pluripotent stem cell-derived neural cells from the basic research tool to a treatment option in regenerative medicine have been well recognized. These cells also offer an interesting tool for in vitro models of neuronal networks to be used for drug screening and neurotoxicological studies and for patient/disease specific in vitro models. Here, as aiming to develop a reductionistic in vitro human neuronal network model, we tested whether human embryonic stem cell (hESC)-derived neural cells could be cultured in human cerebrospinal fluid (CSF) in order to better mimic the in vivo conditions. Our results showed that CSF altered the differentiation of hESC-derived neural cells towards glial cells at the expense of neuronal differentiation. The proliferation rate was reduced in CSF cultures. However, even though the use of CSF as the culture medium altered the glial vs. neuronal differentiation rate, the pre-existing spontaneous activity of the neuronal networks persisted throughout the study. These results suggest that it is possible to develop fully human cell and culture-based environments that can further be modified for various in vitro modeling purposes.
Collapse
Affiliation(s)
- Heikki Kiiski
- Critical Care Medicine Research Group, Department of Intensive Care Unit, Tampere University Hospital , FI-33521 Tampere , Finland ; NeuroGroup, Institute of Biomedical Technology/BioMediTech, University of Tampere , FI-33520 Tampere , Finland
| | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Mäkinen M, Joki T, Ylä-Outinen L, Skottman H, Narkilahti S, Aänismaa R. Fluorescent probes as a tool for cell population tracking in spontaneously active neural networks derived from human pluripotent stem cells. J Neurosci Methods 2013; 215:88-96. [PMID: 23473797 DOI: 10.1016/j.jneumeth.2013.02.019] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2012] [Revised: 02/25/2013] [Accepted: 02/27/2013] [Indexed: 11/19/2022]
Abstract
Applications such as 3D cultures and tissue modelling require cell tracking with non-invasive methods. In this work, the suitability of two fluorescent probes, CellTracker, CT, and long chain carbocyanine dye, DiD, was investigated for long-term culturing of labeled human pluripotent stem cell-derived neural cells. We found that these dyes did not affect the cell viability. However, proliferation was decreased in DiD labeled cell population. With both dyes the labeling was stable up to 4 weeks. CT and DiD labeled cells could be co-cultured and, importantly, these mixed populations had their normal ability to form spontaneous electrical network activity. In conclusion, human neural cells can be successfully labeled with these two fluorescent probes without significantly affecting the cell characteristics. These labeled cells could be utilized further in e.g. building controlled neuronal networks for neurotoxicity screening platforms, combining cells with biomaterials for 3D studies, and graft development.
Collapse
Affiliation(s)
- M Mäkinen
- NeuroGroup, Institute of Biomedical Technology/BioMediTech, Biokatu 12, FI-33014 University of Tampere, Finland.
| | | | | | | | | | | |
Collapse
|
42
|
Toivonen S, Ojala M, Hyysalo A, Ilmarinen T, Rajala K, Pekkanen-Mattila M, Äänismaa R, Lundin K, Palgi J, Weltner J, Trokovic R, Silvennoinen O, Skottman H, Narkilahti S, Aalto-Setälä K, Otonkoski T. Comparative analysis of targeted differentiation of human induced pluripotent stem cells (hiPSCs) and human embryonic stem cells reveals variability associated with incomplete transgene silencing in retrovirally derived hiPSC lines. Stem Cells Transl Med 2013; 2:83-93. [PMID: 23341440 DOI: 10.5966/sctm.2012-0047] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Functional hepatocytes, cardiomyocytes, neurons, and retinal pigment epithelial (RPE) cells derived from human embryonic stem cells (hESCs) or human induced pluripotent stem cells (hiPSCs) could provide a defined and renewable source of human cells relevant for cell replacement therapies, drug discovery, toxicology testing, and disease modeling. In this study, we investigated the differences between the differentiation potentials of three hESC lines, four retrovirally derived hiPSC lines, and one hiPSC line derived with the nonintegrating Sendai virus technology. Four independent protocols were used for hepatocyte, cardiomyocyte, neuronal, and RPE cell differentiation. Overall, cells differentiated from hESCs and hiPSCs showed functional similarities and similar expression of genes characteristic of specific cell types, and differences between individual cell lines were also detected. Reactivation of transgenic OCT4 was detected specifically during RPE differentiation in the retrovirally derived lines, which may have affected the outcome of differentiation with these hiPSCs. One of the hiPSC lines was inferior in all directions, and it failed to produce hepatocytes. Exogenous KLF4 was incompletely silenced in this cell line. No transgene expression was detected in the Sendai virus-derived hiPSC line. These findings highlight the problems related to transgene expression in retrovirally derived hiPSC lines.
Collapse
Affiliation(s)
- Sanna Toivonen
- Research Programs Unit, Molecular Neurology, Biomedicum Stem Cell Center, University of Helsinki, Helsinki, Finland
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Walter J, Dihné M. Species-dependent differences of embryonic stem cell-derived neural stem cells after Interferon gamma treatment. Front Cell Neurosci 2012; 6:52. [PMID: 23162429 PMCID: PMC3492763 DOI: 10.3389/fncel.2012.00052] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2012] [Accepted: 10/17/2012] [Indexed: 12/23/2022] Open
Abstract
Pluripotent stem cell (pSC)-derived, neural stem cells (NSCs) are actually extensively explored in the field of neuroregeneration and to clarify disease mechanisms or model neurological diseases in vitro. Regarding the latter, proliferation and differentiation of pSC-derived NSCs are investigated under the influence of a variety of different substances among them key players of inflammation. However, results generated on a murine genetic background are not always representative for the human situation which increasingly leads to the application of human cell culture systems derived from human pSCs. We investigated here, if the recently described interferon gamma (IFNγ)-induced dysregulated neural phenotype characterized by simultaneous expression of glial and neuronal markers on murine NSCs (Walter et al., 2011, 2012) can also be found on a human genetic background. For this purpose, we performed experiments with human embryonic stem cell-derived NSCs. We could show that the IFNγ-induced dysregulated neural phenotype cannot be induced in human NSCs. This difference occurs, although typical genes like signal transducers and activators of transcription 1 (Stat 1) or interferon regulatory factor 9 (IRF-9) are similarly regulated by IFNγ in both, murine and human populations. These results illustrate that fundamental differences between murine and human neural populations exist in vitro, independent of anatomical system-related properties.
Collapse
Affiliation(s)
- Janine Walter
- Department of Neurology, Heinrich-Heine-University Düsseldorf Düsseldorf, Germany ; Department of Neurology and Epileptology, Hertie Institute for Clinical Brain Research, Eberhard-Karls-University Tübingen, Germany
| | | |
Collapse
|
44
|
Kapucu FE, Tanskanen JMA, Mikkonen JE, Ylä-Outinen L, Narkilahti S, Hyttinen JAK. Burst analysis tool for developing neuronal networks exhibiting highly varying action potential dynamics. Front Comput Neurosci 2012; 6:38. [PMID: 22723778 PMCID: PMC3378047 DOI: 10.3389/fncom.2012.00038] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2012] [Accepted: 05/30/2012] [Indexed: 01/15/2023] Open
Abstract
In this paper we propose a firing statistics based neuronal network burst detection algorithm for neuronal networks exhibiting highly variable action potential dynamics. Electrical activity of neuronal networks is generally analyzed by the occurrences of spikes and bursts both in time and space. Commonly accepted analysis tools employ burst detection algorithms based on predefined criteria. However, maturing neuronal networks, such as those originating from human embryonic stem cells (hESCs), exhibit highly variable network structure and time-varying dynamics. To explore the developing burst/spike activities of such networks, we propose a burst detection algorithm which utilizes the firing statistics based on interspike interval (ISI) histograms. Moreover, the algorithm calculates ISI thresholds for burst spikes as well as for pre-burst spikes and burst tails by evaluating the cumulative moving average (CMA) and skewness of the ISI histogram. Because of the adaptive nature of the proposed algorithm, its analysis power is not limited by the type of neuronal cell network at hand. We demonstrate the functionality of our algorithm with two different types of microelectrode array (MEA) data recorded from spontaneously active hESC-derived neuronal cell networks. The same data was also analyzed by two commonly employed burst detection algorithms and the differences in burst detection results are illustrated. The results demonstrate that our method is both adaptive to the firing statistics of the network and yields successful burst detection from the data. In conclusion, the proposed method is a potential tool for analyzing of hESC-derived neuronal cell networks and thus can be utilized in studies aiming to understand the development and functioning of human neuronal networks and as an analysis tool for in vitro drug screening and neurotoxicity assays.
Collapse
Affiliation(s)
- Fikret E Kapucu
- Department of Biomedical Engineering, Tampere University of Technology Tampere, Finland
| | | | | | | | | | | |
Collapse
|
45
|
Ylä-Outinen L, Joki T, Varjola M, Skottman H, Narkilahti S. Three-dimensional growth matrix for human embryonic stem cell-derived neuronal cells. J Tissue Eng Regen Med 2012; 8:186-94. [PMID: 22611014 DOI: 10.1002/term.1512] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2011] [Revised: 01/30/2012] [Accepted: 02/04/2012] [Indexed: 12/21/2022]
Abstract
The future of tissue engineering applications for neuronal cells will require a supportive 3D matrix. This particular matrix should be soft, elastic and supportive for cell growth. In this study, we characterized the suitability of a 3D synthetic hydrogel matrix, PuraMatrix™, as a growth platform for human embryonic stem cell (hESC)-derived neural cells. The viability of the cells grown on top of, inside and under the hydrogel was monitored. The maturation and electrical activity of the neuronal networks inside the hydrogel were further characterized. We showed that cells stayed viable on the top of the PuraMatrix™ surface and growth of the neural cells and neural processes was good. Further, hESC-derived neurons, astrocytes and oligodendrocytes all grew, matured and migrated when cultured inside the hydrogel. Importantly, neuronal cells were able to form electrically active connections that were verified using microelectrode array. Thus, PuraMatrix is a good supportive growth matrix for human neural cells and may serve as a matrix for neuronal scaffolds in neural tissue engineering.
Collapse
Affiliation(s)
- Laura Ylä-Outinen
- NeuroGroup, Institute of Biomedical Technology, University of Tampere, Tampere, Finland; BioMediTech, Tampere, Finland; The Science Center of Pirkanmaa Hospital District, Tampere University Hospital, Tampere, Finland
| | | | | | | | | |
Collapse
|
46
|
Sison-Young RLC, Kia R, Heslop J, Kelly L, Rowe C, Cross MJ, Kitteringham NR, Hanley N, Park BK, Goldring CEP. Human pluripotent stem cells for modeling toxicity. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2012; 63:207-256. [PMID: 22776643 DOI: 10.1016/b978-0-12-398339-8.00006-9] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/01/2023]
Abstract
The development of xenobiotics, driven by the demand for therapeutic, domestic and industrial uses continues to grow. However, along with this increasing demand is the risk of xenobiotic-induced toxicity. Currently, safety screening of xenobiotics uses a plethora of animal and in vitro model systems which have over the decades proven useful during compound development and for application in mechanistic studies of xenobiotic-induced toxicity. However, these assessments have proven to be animal-intensive and costly. More importantly, the prevalence of xenobiotic-induced toxicity is still significantly high, causing patient morbidity and mortality, and a costly impediment during drug development. This suggests that the current models for drug safety screening are not reliable in toxicity prediction, and the results not easily translatable to the clinic due to insensitive assays that do not recapitulate fully the complex phenotype of a functional cell type in vivo. Recent advances in the field of stem cell research have potentially allowed for a readily available source of metabolically competent cells for toxicity studies, derived using human pluripotent stem cells harnessed from embryos or reprogrammed from mature somatic cells. Pluripotent stem cell-derived cell types also allow for potential disease modeling in vitro for the purposes of drug toxicology and safety pharmacology, making this model possibly more predictive of drug toxicity compared with existing models. This article will review the advances and challenges of using human pluripotent stem cells for modeling metabolism and toxicity, and offer some perspectives as to where its future may lie.
Collapse
Affiliation(s)
- R L C Sison-Young
- MRC Centre for Drug Safety Science, Department of Molecular and Clinical Pharmacology, University of Liverpool, Liverpool, United Kingdom
| | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Aanismaa R, Hautala J, Vuorinen A, Miettinen S, Narkilahti S. Human dental pulp stem cells differentiate into neural precursors but not into mature functional neurons. ACTA ACUST UNITED AC 2012. [DOI: 10.4236/scd.2012.23013] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
48
|
All Titanium Microelectrode Array for Field Potential Measurements from Neurons and Cardiomyocytes—A Feasibility Study. MICROMACHINES 2011. [DOI: 10.3390/mi2040394] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
49
|
Koskela JE, Turunen S, Ylä-Outinen L, Narkilahti S, Kellomäki M. Two-photon microfabrication of poly(ethylene glycol) diacrylate and a novel biodegradable photopolymer-comparison of processability for biomedical applications. POLYM ADVAN TECHNOL 2011. [DOI: 10.1002/pat.2002] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Affiliation(s)
- Jenni E. Koskela
- Tampere University of Technology; Department of Biomedical Engineering; PO Box 692 33101 Tampere Finland
| | - Sanna Turunen
- Tampere University of Technology; Department of Biomedical Engineering; PO Box 692 33101 Tampere Finland
| | - Laura Ylä-Outinen
- Regea-Institute for Regenerative Medicine; University of Tampere and Tampere University Hospital; Biokatu 12 33520 Tampere Finland
- The Science Center of Pirkanmaa Hospital District; Tampere Finland
| | - Susanna Narkilahti
- Regea-Institute for Regenerative Medicine; University of Tampere and Tampere University Hospital; Biokatu 12 33520 Tampere Finland
| | - Minna Kellomäki
- Tampere University of Technology; Department of Biomedical Engineering; PO Box 692 33101 Tampere Finland
| |
Collapse
|
50
|
Ylä-Outinen L, Heikkilä J, Skottman H, Suuronen R, Aänismaa R, Narkilahti S. Human cell-based micro electrode array platform for studying neurotoxicity. FRONTIERS IN NEUROENGINEERING 2010; 3. [PMID: 20953240 PMCID: PMC2955435 DOI: 10.3389/fneng.2010.00111] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/28/2010] [Accepted: 08/30/2010] [Indexed: 12/15/2022]
Abstract
At present, most of the neurotoxicological analyses are based on in vitro and in vivo models utilizing animal cells or animal models. In addition, the used in vitro models are mostly based on molecular biological end-point analyses. Thus, for neurotoxicological screening, human cell-based analysis platforms in which the functional neuronal networks responses for various neurotoxicants can be also detected real-time are highly needed. Microelectrode array (MEA) is a method which enables the measurement of functional activity of neuronal cell networks in vitro for long periods of time. Here, we utilize MEA to study the neurotoxicity of methyl mercury chloride (MeHgCl, concentrations 0.5–500 nM) to human embryonic stem cell (hESC)-derived neuronal cell networks exhibiting spontaneous electrical activity. The neuronal cell cultures were matured on MEAs into networks expressing spontaneous spike train-like activity before exposing the cells to MeHgCl for 72 h. MEA measurements were performed acutely and 24, 48, and 72 h after the onset of the exposure. Finally, exposed cells were analyzed with traditional molecular biological methods for cell proliferation, cell survival, and gene and protein expression. Our results show that 500 nM MeHgCl decreases the electrical signaling and alters the pharmacologic response of hESC-derived neuronal networks in delayed manner whereas effects can not be detected with qRT-PCR, immunostainings, or proliferation measurements. Thus, we conclude that human cell-based MEA platform is a sensitive online method for neurotoxicological screening.
Collapse
Affiliation(s)
- Laura Ylä-Outinen
- Regea - Institute for Regenerative Medicine, University of Tampere and Tampere University Hospital Tampere, Finland
| | | | | | | | | | | |
Collapse
|