1
|
Hong S, Lee J, Kim Y, Kim E, Shin K. AAVS1-targeted, stable expression of ChR2 in human brain organoids for consistent optogenetic control. Bioeng Transl Med 2024; 9:e10690. [PMID: 39545087 PMCID: PMC11558186 DOI: 10.1002/btm2.10690] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Revised: 04/30/2024] [Accepted: 05/23/2024] [Indexed: 11/17/2024] Open
Abstract
Self-organizing brain organoids provide a promising tool for studying human development and disease. Here we created human forebrain organoids with stable and homogeneous expression of channelrhodopsin-2 (ChR2) by generating AAVS1 safe harbor locus-targeted, ChR2 knocked-in human pluripotent stem cells (hPSCs), followed by the differentiation of these genetically engineered hPSCs into forebrain organoids. The resulting ChR2-expressing human forebrain organoids showed homogeneous cellular expression of ChR2 throughout entire regions without any structural and functional perturbations and displayed consistent and robust neural activation upon light stimulation, allowing for the non-virus mediated, spatiotemporal optogenetic control of neural activities. Furthermore, in the hybrid platform in which brain organoids are connected with spinal cord organoids and skeletal muscle spheroids, ChR2 knocked-in forebrain organoids induced strong and consistent muscle contraction upon brain-specific optogenetic stimulation. Our study thus provides a novel, non-virus mediated, preclinical human organoid system for light-inducible, consistent control of neural activities to study neural circuits and dynamics in normal and disease-specific human brains as well as neural connections between brain and other peripheral tissues.
Collapse
Affiliation(s)
- Soojung Hong
- School of Biological Sciences, College of Natural Sciences, Seoul National UniversitySeoulRepublic of Korea
- Institute of Molecular Biology and Genetics, Seoul National UniversitySeoulRepublic of Korea
| | - Juhee Lee
- Institute of Molecular Biology and Genetics, Seoul National UniversitySeoulRepublic of Korea
| | - Yunhee Kim
- School of Biological Sciences, College of Natural Sciences, Seoul National UniversitySeoulRepublic of Korea
- Institute of Molecular Biology and Genetics, Seoul National UniversitySeoulRepublic of Korea
| | - Eunjee Kim
- Institute of Molecular Biology and Genetics, Seoul National UniversitySeoulRepublic of Korea
| | - Kunyoo Shin
- School of Biological Sciences, College of Natural Sciences, Seoul National UniversitySeoulRepublic of Korea
- Institute of Molecular Biology and Genetics, Seoul National UniversitySeoulRepublic of Korea
| |
Collapse
|
2
|
Tang F, Yan F, Zhong Y, Li J, Gong H, Li X. Optogenetic Brain-Computer Interfaces. Bioengineering (Basel) 2024; 11:821. [PMID: 39199779 PMCID: PMC11351350 DOI: 10.3390/bioengineering11080821] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Revised: 07/23/2024] [Accepted: 07/24/2024] [Indexed: 09/01/2024] Open
Abstract
The brain-computer interface (BCI) is one of the most powerful tools in neuroscience and generally includes a recording system, a processor system, and a stimulation system. Optogenetics has the advantages of bidirectional regulation, high spatiotemporal resolution, and cell-specific regulation, which expands the application scenarios of BCIs. In recent years, optogenetic BCIs have become widely used in the lab with the development of materials and software. The systems were designed to be more integrated, lightweight, biocompatible, and power efficient, as were the wireless transmission and chip-level embedded BCIs. The software is also constantly improving, with better real-time performance and accuracy and lower power consumption. On the other hand, as a cutting-edge technology spanning multidisciplinary fields including molecular biology, neuroscience, material engineering, and information processing, optogenetic BCIs have great application potential in neural decoding, enhancing brain function, and treating neural diseases. Here, we review the development and application of optogenetic BCIs. In the future, combined with other functional imaging techniques such as near-infrared spectroscopy (fNIRS) and functional magnetic resonance imaging (fMRI), optogenetic BCIs can modulate the function of specific circuits, facilitate neurological rehabilitation, assist perception, establish a brain-to-brain interface, and be applied in wider application scenarios.
Collapse
Affiliation(s)
- Feifang Tang
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics, MoE Key Laboratory for Biomedical Photonics, Huazhong University of Science and Technology, Wuhan 430074, China; (F.T.); (F.Y.); (Y.Z.); (J.L.); (H.G.)
| | - Feiyang Yan
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics, MoE Key Laboratory for Biomedical Photonics, Huazhong University of Science and Technology, Wuhan 430074, China; (F.T.); (F.Y.); (Y.Z.); (J.L.); (H.G.)
| | - Yushan Zhong
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics, MoE Key Laboratory for Biomedical Photonics, Huazhong University of Science and Technology, Wuhan 430074, China; (F.T.); (F.Y.); (Y.Z.); (J.L.); (H.G.)
| | - Jinqian Li
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics, MoE Key Laboratory for Biomedical Photonics, Huazhong University of Science and Technology, Wuhan 430074, China; (F.T.); (F.Y.); (Y.Z.); (J.L.); (H.G.)
| | - Hui Gong
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics, MoE Key Laboratory for Biomedical Photonics, Huazhong University of Science and Technology, Wuhan 430074, China; (F.T.); (F.Y.); (Y.Z.); (J.L.); (H.G.)
| | - Xiangning Li
- Key Laboratory of Biomedical Engineering of Hainan Province, School of Biomedical Engineering, Hainan University, Haikou 570228, China
| |
Collapse
|
3
|
Martinez-Curiel R, Jansson L, Tsupykov O, Avaliani N, Aretio-Medina C, Hidalgo I, Monni E, Bengzon J, Skibo G, Lindvall O, Kokaia Z, Palma-Tortosa S. Oligodendrocytes in human induced pluripotent stem cell-derived cortical grafts remyelinate adult rat and human cortical neurons. Stem Cell Reports 2023; 18:1643-1656. [PMID: 37236198 PMCID: PMC10444570 DOI: 10.1016/j.stemcr.2023.04.010] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2022] [Revised: 04/24/2023] [Accepted: 04/25/2023] [Indexed: 05/28/2023] Open
Abstract
Neuronal loss and axonal demyelination underlie long-term functional impairments in patients affected by brain disorders such as ischemic stroke. Stem cell-based approaches reconstructing and remyelinating brain neural circuitry, leading to recovery, are highly warranted. Here, we demonstrate the in vitro and in vivo production of myelinating oligodendrocytes from a human induced pluripotent stem cell (iPSC)-derived long-term neuroepithelial stem (lt-NES) cell line, which also gives rise to neurons with the capacity to integrate into stroke-injured, adult rat cortical networks. Most importantly, the generated oligodendrocytes survive and form myelin-ensheathing human axons in the host tissue after grafting onto adult human cortical organotypic cultures. This lt-NES cell line is the first human stem cell source that, after intracerebral delivery, can repair both injured neural circuitries and demyelinated axons. Our findings provide supportive evidence for the potential future use of human iPSC-derived cell lines to promote effective clinical recovery following brain injuries.
Collapse
Affiliation(s)
- Raquel Martinez-Curiel
- Laboratory of Stem Cells and Restorative Neurology, Lund Stem Cell Center, Lund University, 22184 Lund, Sweden
| | - Linda Jansson
- Laboratory of Stem Cells and Restorative Neurology, Lund Stem Cell Center, Lund University, 22184 Lund, Sweden
| | - Oleg Tsupykov
- Department of Cytology, Bogomoletz Institute of Physiology; Institute of Genetic and Regenerative Medicine, Strazhesko National Scientific Center of Cardiology, Clinical and Regenerative Medicine, 01024 Kyiv, Ukraine
| | | | - Constanza Aretio-Medina
- Laboratory of Stem Cells and Restorative Neurology, Lund Stem Cell Center, Lund University, 22184 Lund, Sweden
| | - Isabel Hidalgo
- Division of Molecular Hematology, Wallenberg Center for Molecular Medicine, Lund Stem Cell Center, Lund University, 22184 Lund, Sweden
| | - Emanuela Monni
- Lund Stem Cell Center, Lund University, 22184 Lund, Sweden
| | - Johan Bengzon
- Division of Neurosurgery, Department of Clinical Sciences Lund, University Hospital, 22184 Lund, Sweden
| | - Galyna Skibo
- Department of Cytology, Bogomoletz Institute of Physiology; Institute of Genetic and Regenerative Medicine, Strazhesko National Scientific Center of Cardiology, Clinical and Regenerative Medicine, 01024 Kyiv, Ukraine
| | - Olle Lindvall
- Laboratory of Stem Cells and Restorative Neurology, Lund Stem Cell Center, Lund University, 22184 Lund, Sweden
| | - Zaal Kokaia
- Laboratory of Stem Cells and Restorative Neurology, Lund Stem Cell Center, Lund University, 22184 Lund, Sweden.
| | - Sara Palma-Tortosa
- Laboratory of Stem Cells and Restorative Neurology, Lund Stem Cell Center, Lund University, 22184 Lund, Sweden
| |
Collapse
|
4
|
Ghuman H, Perry N, Grice L, Gerwig M, Moorhead J, Nitzsche F, Poplawsky AJ, Ambrosio F, Modo M. Physical therapy exerts sub-additive and suppressive effects on intracerebral neural stem cell implantation in a rat model of stroke. J Cereb Blood Flow Metab 2022; 42:826-843. [PMID: 34826373 PMCID: PMC9254031 DOI: 10.1177/0271678x211062955] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Intracerebral cell therapy (CT) is emerging as a new therapeutic paradigm for stroke. However, the impact of physical therapy (PT) on implanted cells and their ability to promote recovery remains poorly understood. To address this translational issue, a clinical-grade neural stem cell (NSC) line was implanted into peri-infarct tissue using MRI-defined injection sites, two weeks after stroke. PT in the form of aerobic exercise (AE) was administered 5 × per week post-implantation using a paradigm commonly applied in patients with stroke. A combined AE and CT exerted sub-additive therapeutic effects on sensory neglect, whereas AE suppressed CT effects on motor integration and grip strength. Behavioral testing emerged as a potentially major component for task integration. It is expected that this study will guide and inform the incorporation of PT in the design of clinical trials evaluating intraparenchymal NSCs implantation for stroke.
Collapse
Affiliation(s)
- Harmanvir Ghuman
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.,Department of Bioengineering, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Nikhita Perry
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Lauren Grice
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Madeline Gerwig
- Department of Neuroscience, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Jeffrey Moorhead
- Department of Physical Medicine and Rehabilitation, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.,Department of Radiology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Franziska Nitzsche
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.,Department of Radiology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | | | - Fabrisia Ambrosio
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.,Department of Bioengineering, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.,Department of Physical Medicine and Rehabilitation, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Michel Modo
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.,Department of Bioengineering, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.,Department of Radiology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
5
|
Ceto S, Courtine G. Optogenetic Interrogation of Circuits Following Neurotrauma. Front Mol Neurosci 2022; 14:803856. [PMID: 34975403 PMCID: PMC8716760 DOI: 10.3389/fnmol.2021.803856] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Accepted: 11/18/2021] [Indexed: 11/13/2022] Open
Abstract
Biological and engineering strategies for neural repair and recovery from neurotrauma continue to emerge at a rapid pace. Until recently, studies of the impact of neurotrauma and repair strategies on the reorganization of the central nervous system have focused on broadly defined circuits and pathways. Optogenetic modulation and recording methods now enable the interrogation of precisely defined neuronal populations in the brain and spinal cord, allowing unprecedented precision in electrophysiological and behavioral experiments. This mini-review summarizes the spectrum of light-based tools that are currently available to probe the properties and functions of well-defined neuronal subpopulations in the context of neurotrauma. In particular, we highlight the challenges to implement these tools in damaged and reorganizing tissues, and we discuss best practices to overcome these obstacles.
Collapse
Affiliation(s)
- Steven Ceto
- Center for Neuroprosthetics and Brain Mind Institute, School of Life Sciences, Swiss Federal Institute of Technology (EPFL), Lausanne, Switzerland.,Department of Clinical Neuroscience, Lausanne University Hospital (CHUV) and University of Lausanne (UNIL), Lausanne, Switzerland.,Defitech Center for Interventional Neurotherapies (.NeuroRestore), EPFL/CHUV/UNIL, Lausanne, Switzerland
| | - Grégoire Courtine
- Center for Neuroprosthetics and Brain Mind Institute, School of Life Sciences, Swiss Federal Institute of Technology (EPFL), Lausanne, Switzerland.,Department of Clinical Neuroscience, Lausanne University Hospital (CHUV) and University of Lausanne (UNIL), Lausanne, Switzerland.,Defitech Center for Interventional Neurotherapies (.NeuroRestore), EPFL/CHUV/UNIL, Lausanne, Switzerland.,Department of Neurosurgery, Lausanne University Hospital (CHUV), Lausanne, Switzerland
| |
Collapse
|
6
|
Human stem cell-derived GABAergic neurons functionally integrate into human neuronal networks. Sci Rep 2021; 11:22050. [PMID: 34764308 PMCID: PMC8585944 DOI: 10.1038/s41598-021-01270-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Accepted: 10/26/2021] [Indexed: 02/03/2023] Open
Abstract
Gamma-aminobutyric acid (GABA)-releasing interneurons modulate neuronal network activity in the brain by inhibiting other neurons. The alteration or absence of these cells disrupts the balance between excitatory and inhibitory processes, leading to neurological disorders such as epilepsy. In this regard, cell-based therapy may be an alternative therapeutic approach. We generated light-sensitive human embryonic stem cell (hESC)-derived GABAergic interneurons (hdIN) and tested their functionality. After 35 days in vitro (DIV), hdINs showed electrophysiological properties and spontaneous synaptic currents comparable to mature neurons. In co-culture with human cortical neurons and after transplantation (AT) into human brain tissue resected from patients with drug-resistant epilepsy, light-activated channelrhodopsin-2 (ChR2) expressing hdINs induced postsynaptic currents in human neurons, strongly suggesting functional efferent synapse formation. These results provide a proof-of-concept that hESC-derived neurons can integrate and modulate the activity of a human host neuronal network. Therefore, this study supports the possibility of precise temporal control of network excitability by transplantation of light-sensitive interneurons.
Collapse
|
7
|
Novel Approaches Used to Examine and Control Neurogenesis in Parkinson's Disease. Int J Mol Sci 2021; 22:ijms22179608. [PMID: 34502516 PMCID: PMC8431772 DOI: 10.3390/ijms22179608] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Revised: 08/31/2021] [Accepted: 09/02/2021] [Indexed: 12/16/2022] Open
Abstract
Neurogenesis is a key mechanism of brain development and plasticity, which is impaired in chronic neurodegeneration, including Parkinson’s disease. The accumulation of aberrant α-synuclein is one of the features of PD. Being secreted, this protein produces a prominent neurotoxic effect, alters synaptic plasticity, deregulates intercellular communication, and supports the development of neuroinflammation, thereby providing propagation of pathological events leading to the establishment of a PD-specific phenotype. Multidirectional and ambiguous effects of α-synuclein on adult neurogenesis suggest that impaired neurogenesis should be considered as a target for the prevention of cell loss and restoration of neurological functions. Thus, stimulation of endogenous neurogenesis or cell-replacement therapy with stem cell-derived differentiated neurons raises new hopes for the development of effective and safe technologies for treating PD neurodegeneration. Given the rapid development of optogenetics, it is not surprising that this method has already been repeatedly tested in manipulating neurogenesis in vivo and in vitro via targeting stem or progenitor cells. However, niche astrocytes could also serve as promising candidates for controlling neuronal differentiation and improving the functional integration of newly formed neurons within the brain tissue. In this review, we mainly focus on current approaches to assess neurogenesis and prospects in the application of optogenetic protocols to restore the neurogenesis in Parkinson’s disease.
Collapse
|
8
|
Kamaraj A, Kyriacou H, Seah KTM, Khan WS. Use of human induced pluripotent stem cells for cartilage regeneration in vitro and within chondral defect models of knee joint cartilage in vivo: a Preferred Reporting Items for Systematic Reviews and Meta-Analyses systematic literature review. Cytotherapy 2021; 23:647-661. [PMID: 34059422 DOI: 10.1016/j.jcyt.2021.03.008] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 03/16/2021] [Accepted: 03/27/2021] [Indexed: 12/16/2022]
Abstract
BACKGROUND AIMS Articular cartilage has limited regenerative ability when damaged through trauma or disease. Failure to treat focal chondral lesions results in changes that inevitably progress to osteoarthritis. Osteoarthritis is a major contributor to disability globally, which results in significant medical costs and lost wages every year. Human induced pluripotent stem cells (hiPSCs) have long been considered a potential autologous therapeutic option for the treatment of focal chondral lesions. Although there are significant advantages to hiPSCs over other stem cell options, such as mesenchymal and embryonic stem cells, there are concerns regarding their ability to form bona fide cartilage and their tumorgenicity in vivo. METHODS The authors carried out a systematic literature review on the use of hiPSCs to produce differentiated progeny capable of producing high-quality cartilage in vitro and regenerate cartilage in osteochondral defects in vivo in accordance with Preferred Reporting Items for Systematic Reviews and Meta-Analyses guidelines. Eight studies were included in the review that used hiPSCs or their derived progeny in xenogeneic transplants in animal models to regenerate cartilage in osteochondral defects of the knee joint. The in vitro-differentiated, hiPSC-derived and in vivo defect repair ability of the hiPSC-derived progeny transplants were assessed. RESULTS Most studies reported the generation of high-quality cartilage-producing progeny that were able to successfully repair cartilage defects in vivo. No tumorigenicity was observed. CONCLUSIONS The authors conclude that hiPSCs offer a valuable source of cartilage-producing progeny that show promise as an effective cell-based therapy in treating focal chondral lesions.
Collapse
Affiliation(s)
- Achi Kamaraj
- Division of Trauma and Orthopedic Surgery, Addenbrooke's Hospital, University of Cambridge, Cambridge, UK
| | - Harry Kyriacou
- Division of Trauma and Orthopedic Surgery, Addenbrooke's Hospital, University of Cambridge, Cambridge, UK
| | - K T Matthew Seah
- Division of Trauma and Orthopedic Surgery, Addenbrooke's Hospital, University of Cambridge, Cambridge, UK.
| | - Wasim S Khan
- Division of Trauma and Orthopedic Surgery, Addenbrooke's Hospital, University of Cambridge, Cambridge, UK
| |
Collapse
|
9
|
Butler-Ryan R, Wood IC. Efficient infection of organotypic hippocampal slice cultures with adenovirus carrying the transgene REST/NRSF. J Neurosci Methods 2021; 356:109147. [PMID: 33771654 DOI: 10.1016/j.jneumeth.2021.109147] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Revised: 02/24/2021] [Accepted: 03/15/2021] [Indexed: 11/29/2022]
Abstract
Organotypic hippocampal slice cultures provide a useful platform maintaining hippocampal structure and synaptic connections of the brain over weeks in culture with ease of in vitro manipulations. Gene transfer is a particularly desirable tool for using with them but current difficulties with transformation of transgenes into these cultures is a barrier to their use in research. Previous quantifications of viral infections have shown low transformation rates and have relied upon invasive microinjections. In this paper we present an efficient way of infecting organotypic cultures with adenovirus at the acute slice stage that does not require injection. We use the adenoviral delivery system to introduce the transcription factor REST and a GFP marker, providing around 41 % cellular infection spread throughout the entire slice culture and promoting transgene expression for weeks in vitro. GFP expression was observed most intensely in the slices when they were infected just a few hours after plating and was shown to infect neurons and microglia. We decided to use the transcription factor REST/NRSF as an example transgene which was delivered into cells via the adenoviral construct, conferring overexpression of REST in addition to the GFP marker. This outlines a technique whereby adenoviral infection of organotypic cultures can infect neurons with good efficiency and confer successful manipulation of genetic factors within the cell.
Collapse
Affiliation(s)
- Ruth Butler-Ryan
- School of Biomedical Sciences, Faculty of Biological Sciences, University of Leeds, Leeds, LS2 9JT, UK.
| | - Ian C Wood
- School of Biomedical Sciences, Faculty of Biological Sciences, University of Leeds, Leeds, LS2 9JT, UK.
| |
Collapse
|
10
|
Asano T, Teh DBL, Yawo H. Application of Optogenetics for Muscle Cells and Stem Cells. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1293:359-375. [PMID: 33398826 DOI: 10.1007/978-981-15-8763-4_23] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
This chapter describes the current progress of basic research, and potential therapeutic applications primarily focused on the optical manipulation of muscle cells and neural stem cells using microbial rhodopsin as a light-sensitive molecule. Since the contractions of skeletal, cardiac, and smooth muscle cells are mainly regulated through their membrane potential, several studies have been demonstrated to up- or downregulate the muscle contraction directly or indirectly using optogenetic actuators or silencers with defined stimulation patterns and intensities. Light-dependent oscillation of membrane potential also facilitates the maturation of myocytes with the development of T tubules and sarcomere structures, tandem arrays of minimum contractile units consists of contractile proteins and cytoskeletal proteins. Optogenetics has been applied to various stem cells and multipotent/pluripotent cells such as embryonic stem cells (ESCs) and induced pluripotent stem cells (iPSCs) to generate light-sensitive neurons and to facilitate neuroscience. The chronic optical stimulation of the channelrhodopsin-expressing neural stem cells facilitates their neural differentiation. There are potential therapeutic applications of optogenetics in cardiac pacemaking, muscle regeneration/maintenance, locomotion recovery for the treatment of muscle paralysis due to motor neuron diseases such as amyotrophic lateral sclerosis (ALS). Optogenetics would also facilitate maturation, network integration of grafted neurons, and improve the microenvironment around them when applied to stem cells.
Collapse
Affiliation(s)
- Toshifumi Asano
- Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Bunkyo-ku, Tokyo, Japan
| | - Daniel Boon Loong Teh
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Hiromu Yawo
- The Institute for Solid State Physics, The University of Tokyo, Kashiwa, Japan.
| |
Collapse
|
11
|
Hirose S, Tanaka Y, Shibata M, Kimura Y, Ishikawa M, Higurashi N, Yamamoto T, Ichise E, Chiyonobu T, Ishii A. Application of induced pluripotent stem cells in epilepsy. Mol Cell Neurosci 2020; 108:103535. [DOI: 10.1016/j.mcn.2020.103535] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Revised: 07/10/2020] [Accepted: 07/31/2020] [Indexed: 02/06/2023] Open
|
12
|
Habibey R, Sharma K, Swiersy A, Busskamp V. Optogenetics for neural transplant manipulation and functional analysis. Biochem Biophys Res Commun 2020; 527:343-349. [PMID: 32033753 DOI: 10.1016/j.bbrc.2020.01.141] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Accepted: 01/24/2020] [Indexed: 01/02/2023]
Abstract
Transplantation of neural stem cells (NSCs) or NSC-derived neurons into the brain is a promising therapeutic approach to restore neuronal function. Rapid progress in the NSCs research field, particularly due to the exploitation of induced pluripotent stem cells (iPSCs), offers great potential and an unlimited source of stem cell-derived neural grafts. Studying the functional integration of these grafts into host brain tissues and their effects on each other have been boosted by the implementation of optogenetic technologies. Optogenetics provides high spatiotemporal functional manipulations of grafted or host neurons in parallel. This review aims to highlight the impact of optogenetics in neural stem cell transplantations.
Collapse
Affiliation(s)
- Rouhollah Habibey
- Technische Universität Dresden, CRTD - Center for Regenerative Therapies at TU Dresden, D-01307, Dresden, Germany
| | - Kritika Sharma
- Technische Universität Dresden, CRTD - Center for Regenerative Therapies at TU Dresden, D-01307, Dresden, Germany
| | - Anka Swiersy
- Technische Universität Dresden, CRTD - Center for Regenerative Therapies at TU Dresden, D-01307, Dresden, Germany
| | - Volker Busskamp
- Technische Universität Dresden, CRTD - Center for Regenerative Therapies at TU Dresden, D-01307, Dresden, Germany; Universitäts-Augenklinik Bonn, University of Bonn, Dep. of Ophthalmology, D-53127, Bonn, Germany.
| |
Collapse
|
13
|
Sterlini B, Fruscione F, Baldassari S, Benfenati F, Zara F, Corradi A. Progress of Induced Pluripotent Stem Cell Technologies to Understand Genetic Epilepsy. Int J Mol Sci 2020; 21:ijms21020482. [PMID: 31940887 PMCID: PMC7013950 DOI: 10.3390/ijms21020482] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Revised: 01/08/2020] [Accepted: 01/10/2020] [Indexed: 12/20/2022] Open
Abstract
The study of the pathomechanisms by which gene mutations lead to neurological diseases has benefit from several cellular and animal models. Recently, induced Pluripotent Stem Cell (iPSC) technologies have made possible the access to human neurons to study nervous system disease-related mechanisms, and are at the forefront of the research into neurological diseases. In this review, we will focalize upon genetic epilepsy, and summarize the most recent studies in which iPSC-based technologies were used to gain insight on the molecular bases of epilepsies. Moreover, we discuss the latest advancements in epilepsy cell modeling. At the two dimensional (2D) level, single-cell models of iPSC-derived neurons lead to a mature neuronal phenotype, and now allow a reliable investigation of synaptic transmission and plasticity. In addition, functional characterization of cerebral organoids enlightens neuronal network dynamics in a three-dimensional (3D) structure. Finally, we discuss the use of iPSCs as the cutting-edge technology for cell therapy in epilepsy.
Collapse
Affiliation(s)
- Bruno Sterlini
- Department of Experimental Medicine, University of Genova, Viale Benedetto XV, 3, 16132 Genoa, Italy;
- Center for Synaptic Neuroscience and Technology, Istituto Italiano di Tecnologia, Largo Rosanna Benzi 10, 16132 Genoa, Italy;
| | - Floriana Fruscione
- Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, University of Genoa, Largo P. Daneo 3, 16132 Genoa, Italy;
| | - Simona Baldassari
- Unità Operativa Complessa Genetica Medica, Istituto di Ricovero e Cura a Carattere Scientifico Giannina Gaslini, Genova Italy, Via G. Gaslini 5, 16147 Genoa, Italy;
| | - Fabio Benfenati
- Center for Synaptic Neuroscience and Technology, Istituto Italiano di Tecnologia, Largo Rosanna Benzi 10, 16132 Genoa, Italy;
- Istituto di Ricovero e Cura a Carattere Scientifico, Ospedale Policlinico San Martino, Largo Rosanna Benzi 10, 16132 Genoa, Italy
| | - Federico Zara
- Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, University of Genoa, Largo P. Daneo 3, 16132 Genoa, Italy;
- Unità Operativa Complessa Genetica Medica, Istituto di Ricovero e Cura a Carattere Scientifico Giannina Gaslini, Genova Italy, Via G. Gaslini 5, 16147 Genoa, Italy;
- Correspondence: (F.Z.); (A.C.)
| | - Anna Corradi
- Department of Experimental Medicine, University of Genova, Viale Benedetto XV, 3, 16132 Genoa, Italy;
- Istituto di Ricovero e Cura a Carattere Scientifico, Ospedale Policlinico San Martino, Largo Rosanna Benzi 10, 16132 Genoa, Italy
- Correspondence: (F.Z.); (A.C.)
| |
Collapse
|
14
|
Immature electrophysiological properties of human-induced pluripotent stem cell-derived neurons transplanted into the mouse cortex for 7 weeks. Neuroreport 2019; 30:169-173. [PMID: 30557202 DOI: 10.1097/wnr.0000000000001178] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The transplantation of human-induced pluripotent stem cell (hiPSC)-derived cells has emerged as a potential clinical approach for the treatment of brain diseases. Recent studies with animal disease models have shown that hiPSC-derived neurons transplanted into the brain, especially the nigrostriatal area, could restore degenerated brain functions. Further works are required to test whether hiPSC-derived neurons can also gain functional properties for other cortical areas. In this study, hiPSC-derived neurospheres were transplanted into the adult mouse hippocampus and sensory cortex. Most transplanted hiPSC-derived neurons expressed both Nestin and NeuN at 7 weeks after transplantation. Whole-cell patch-clamp recordings from brain slices indicated that transplanted cells showed no action potentials upon current injection and few small inward currents, indicating that hiPSC-derived neurons did not become functionally mature within these time periods.
Collapse
|
15
|
Ryu J, Vincent PFY, Ziogas NK, Xu L, Sadeghpour S, Curtin J, Alexandris AS, Stewart N, Sima R, du Lac S, Glowatzki E, Koliatsos VE. Optogenetically transduced human ES cell-derived neural progenitors and their neuronal progenies: Phenotypic characterization and responses to optical stimulation. PLoS One 2019; 14:e0224846. [PMID: 31710637 PMCID: PMC6844486 DOI: 10.1371/journal.pone.0224846] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Accepted: 10/22/2019] [Indexed: 02/04/2023] Open
Abstract
Optogenetically engineered human neural progenitors (hNPs) are viewed as promising tools in regenerative neuroscience because they allow the testing of the ability of hNPs to integrate within nervous system of an appropriate host not only structurally, but also functionally based on the responses of their differentiated progenies to light. Here, we transduced H9 embryonic stem cell-derived hNPs with a lentivirus harboring human channelrhodopsin (hChR2) and differentiated them into a forebrain lineage. We extensively characterized the fate and optogenetic functionality of hChR2-hNPs in vitro with electrophysiology and immunocytochemistry. We also explored whether the in vivo phenotype of ChR2-hNPs conforms to in vitro observations by grafting them into the frontal neocortex of rodents and analyzing their survival and neuronal differentiation. Human ChR2-hNPs acquired neuronal phenotypes (TUJ1, MAP2, SMI-312, and synapsin 1 immunoreactivity) in vitro after an average of 70 days of coculturing with CD1 astrocytes and progressively displayed both inhibitory and excitatory neurotransmitter signatures by immunocytochemistry and whole-cell patch clamp recording. Three months after transplantation into motor cortex of naïve or injured mice, 60–70% of hChR2-hNPs at the transplantation site expressed TUJ1 and had neuronal cytologies, whereas 60% of cells also expressed ChR2. Transplant-derived neurons extended axons through major commissural and descending tracts and issued synaptophysin+ terminals in the claustrum, endopiriform area, and corresponding insular and piriform cortices. There was no apparent difference in engraftment, differentiation, or connectivity patterns between injured and sham subjects. Same trends were observed in a second rodent host, i.e. rat, where we employed longer survival times and found that the majority of grafted hChR2-hNPs differentiated into GABAergic neurons that established dense terminal fields and innervated mostly dendritic profiles in host cortical neurons. In physiological experiments, human ChR2+ neurons in culture generated spontaneous action potentials (APs) 100–170 days into differentiation and their firing activity was consistently driven by optical stimulation. Stimulation generated glutamatergic and GABAergic postsynaptic activity in neighboring ChR2- cells, evidence that hChR2-hNP-derived neurons had established functional synaptic connections with other neurons in culture. Light stimulation of hChR2-hNP transplants in vivo generated complicated results, in part because of the variable response of the transplants themselves. Our findings show that we can successfully derive hNPs with optogenetic properties that are fully transferrable to their differentiated neuronal progenies. We also show that these progenies have substantial neurotransmitter plasticity in vitro, whereas in vivo they mostly differentiate into inhibitory GABAergic neurons. Furthermore, neurons derived from hNPs have the capacity of establishing functional synapses with postsynaptic neurons in vitro, but this outcome is technically challenging to explore in vivo. We propose that optogenetically endowed hNPs hold great promise as tools to explore de novo circuit formation in the brain and, in the future, perhaps launch a new generation of neuromodulatory therapies.
Collapse
Affiliation(s)
- Jiwon Ryu
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Philippe F. Y. Vincent
- Department of Otolaryngology Head and Neck Surgery, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Nikolaos K. Ziogas
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Leyan Xu
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Shirin Sadeghpour
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - John Curtin
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Athanasios S. Alexandris
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Nicholas Stewart
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Richard Sima
- Department of Otolaryngology Head and Neck Surgery, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Sascha du Lac
- Department of Otolaryngology Head and Neck Surgery, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Elisabeth Glowatzki
- Department of Otolaryngology Head and Neck Surgery, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Vassilis E. Koliatsos
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
- Division of Neuropathology, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
- Department of Psychiatry, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
- * E-mail:
| |
Collapse
|
16
|
Sandvig A, Sandvig I. Connectomics of Morphogenetically Engineered Neurons as a Predictor of Functional Integration in the Ischemic Brain. Front Neurol 2019; 10:630. [PMID: 31249553 PMCID: PMC6582372 DOI: 10.3389/fneur.2019.00630] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Accepted: 05/28/2019] [Indexed: 11/13/2022] Open
Abstract
Recent advances in cell reprogramming technologies enable the in vitro generation of theoretically unlimited numbers of cells, including cells of neural lineage and specific neuronal subtypes from human, including patient-specific, somatic cells. Similarly, as demonstrated in recent animal studies, by applying morphogenetic neuroengineering principles in situ, it is possible to reprogram resident brain cells to the desired phenotype. These developments open new exciting possibilities for cell replacement therapy in stroke, albeit not without caveats. Main challenges include the successful integration of engineered cells in the ischemic brain to promote functional restoration as well as the fact that the underlying mechanisms of action are not fully understood. In this review, we aim to provide new insights to the above in the context of connectomics of morphogenetically engineered neural networks. Specifically, we discuss the relevance of combining advanced interdisciplinary approaches to: validate the functionality of engineered neurons by studying their self-organizing behavior into neural networks as well as responses to stroke-related pathology in vitro; derive structural and functional connectomes from these networks in healthy and perturbed conditions; and identify and extract key elements regulating neural network dynamics, which might predict the behavior of grafted engineered neurons post-transplantation in the stroke-injured brain.
Collapse
Affiliation(s)
- Axel Sandvig
- Department of Neuromedicine and Movement Science, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway.,Department of Neurology, St. Olav's Hospital, Trondheim University Hospital, Trondheim, Norway.,Department of Pharmacology and Clinical Neurosciences, Division of Neuro, Head, and Neck, Umeå University Hospital, Umeå, Sweden
| | - Ioanna Sandvig
- Department of Neuromedicine and Movement Science, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway
| |
Collapse
|
17
|
Shiri Z, Simorgh S, Naderi S, Baharvand H. Optogenetics in the Era of Cerebral Organoids. Trends Biotechnol 2019; 37:1282-1294. [PMID: 31227305 DOI: 10.1016/j.tibtech.2019.05.009] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2019] [Revised: 05/18/2019] [Accepted: 05/23/2019] [Indexed: 02/08/2023]
Abstract
The human brain has been deemed the most complex organ and has captivated neuroscientists for decades. Most studies of this organ have relied on reductionist model systems. Although all model systems are essentially wrong, cerebral organoids so far represent the closest recapitulation of human brain development and disease both in terms of cell diversity and organization. The optogenetic technique can be used in this context to study the functional neuroanatomy of the brain, to examine the neural circuits, and to determine the etiology of neurological disorders. In this opinion article, we suggest ways in which optogenetics can be combined with cerebral organoids to allow unprecedented precision and accuracy in studying normal and aberrant neurodevelopmental processes and, as well, neurodegenerative diseases.
Collapse
Affiliation(s)
- Zahra Shiri
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Susan Simorgh
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Somayeh Naderi
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Hossein Baharvand
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran; Department of Developmental Biology, University of Science and Culture, Tehran, Iran.
| |
Collapse
|
18
|
Taoufik E, Kouroupi G, Zygogianni O, Matsas R. Synaptic dysfunction in neurodegenerative and neurodevelopmental diseases: an overview of induced pluripotent stem-cell-based disease models. Open Biol 2019; 8:rsob.180138. [PMID: 30185603 PMCID: PMC6170506 DOI: 10.1098/rsob.180138] [Citation(s) in RCA: 119] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2018] [Accepted: 08/14/2018] [Indexed: 12/12/2022] Open
Abstract
Synaptic dysfunction in CNS disorders is the outcome of perturbations in physiological synapse structure and function, and can be either the cause or the consequence in specific pathologies. Accumulating data in the field of neuropsychiatric disorders, including autism spectrum disorders, schizophrenia and bipolar disorder, point to a neurodevelopmental origin of these pathologies. Due to a relatively early onset of behavioural and cognitive symptoms, it is generally acknowledged that mental illness initiates at the synapse level. On the other hand, synaptic dysfunction has been considered as an endpoint incident in neurodegenerative diseases, such as Alzheimer's, Parkinson's and Huntington's, mainly due to the considerably later onset of clinical symptoms and progressive appearance of cognitive deficits. This dichotomy has recently been challenged, particularly since the discovery of cell reprogramming technologies and the generation of induced pluripotent stem cells from patient somatic cells. The creation of 'disease-in-a-dish' models for multiple CNS pathologies has revealed unexpected commonalities in the molecular and cellular mechanisms operating in both developmental and degenerative conditions, most of which meet at the synapse level. In this review we discuss synaptic dysfunction in prototype neurodevelopmental and neurodegenerative diseases, emphasizing overlapping features of synaptopathy that have been suggested by studies using induced pluripotent stem-cell-based systems. These valuable disease models have highlighted a potential neurodevelopmental component in classical neurodegenerative diseases that is worth pursuing and investigating further. Moving from demonstration of correlation to understanding mechanistic causality forms the basis for developing novel therapeutics.
Collapse
Affiliation(s)
- Era Taoufik
- Laboratory of Cellular and Molecular Neurobiology-Stem Cells, Department of Neurobiology, Hellenic Pasteur Institute, 127 Vassilissis Sofias Avenue, 11521 Athens, Greece
| | - Georgia Kouroupi
- Laboratory of Cellular and Molecular Neurobiology-Stem Cells, Department of Neurobiology, Hellenic Pasteur Institute, 127 Vassilissis Sofias Avenue, 11521 Athens, Greece
| | - Ourania Zygogianni
- Laboratory of Cellular and Molecular Neurobiology-Stem Cells, Department of Neurobiology, Hellenic Pasteur Institute, 127 Vassilissis Sofias Avenue, 11521 Athens, Greece
| | - Rebecca Matsas
- Laboratory of Cellular and Molecular Neurobiology-Stem Cells, Department of Neurobiology, Hellenic Pasteur Institute, 127 Vassilissis Sofias Avenue, 11521 Athens, Greece
| |
Collapse
|
19
|
Duru LN, Quan Z, Qazi TJ, Qing H. Stem cells technology: a powerful tool behind new brain treatments. Drug Deliv Transl Res 2018; 8:1564-1591. [PMID: 29916013 DOI: 10.1007/s13346-018-0548-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Stem cell research has recently become a hot research topic in biomedical research due to the foreseen unlimited potential of stem cells in tissue engineering and regenerative medicine. For many years, medicine has been facing intense challenges, such as an insufficient number of organ donations that is preventing clinicians to fulfill the increasing needs. To try and overcome this regrettable matter, research has been aiming at developing strategies to facilitate the in vitro culture and study of stem cells as a tool for tissue regeneration. Meanwhile, new developments in the microfluidics technology brought forward emerging cell culture applications that are currently allowing for a better chemical and physical control of cellular microenvironment. This review presents the latest developments in stem cell research that brought new therapies to the clinics and how the convergence of the microfluidics technology with stem cell research can have positive outcomes on the fields of regenerative medicine and high-throughput screening. These advances will bring new translational solutions for drug discovery and will upgrade in vitro cell culture to a new level of accuracy and performance. We hope this review will provide new insights into the understanding of new brain treatments from the perspective of stem cell technology especially regarding regenerative medicine and tissue engineering.
Collapse
Affiliation(s)
- Lucienne N Duru
- School of Life Science, Beijing Institute of Technology, Beijing, China
| | - Zhenzhen Quan
- School of Life Science, Beijing Institute of Technology, Beijing, China
| | - Talal Jamil Qazi
- School of Life Science, Beijing Institute of Technology, Beijing, China
| | - Hong Qing
- School of Life Science, Beijing Institute of Technology, Beijing, China. .,Beijing Key Laboratory of Separation and Analysis in Biomedical and Pharmaceuticals, Department of Biomedical Engineering, School of Life Science, Beijing Institute of Technology, 5 South Zhongguancun Street, Haidian District, Beijing, 100081, China.
| |
Collapse
|
20
|
Backofen-Wehrhahn B, Gey L, Bröer S, Petersen B, Schiff M, Handreck A, Stanslowsky N, Scharrenbroich J, Weißing M, Staege S, Wegner F, Niemann H, Löscher W, Gernert M. Anticonvulsant effects after grafting of rat, porcine, and human mesencephalic neural progenitor cells into the rat subthalamic nucleus. Exp Neurol 2018; 310:70-83. [PMID: 30205107 DOI: 10.1016/j.expneurol.2018.09.004] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Revised: 08/20/2018] [Accepted: 09/07/2018] [Indexed: 12/11/2022]
Abstract
Cell transplantation based therapy is a promising strategy for treating intractable epilepsies. Inhibition of the subthalamic nucleus (STN) or substantia nigra pars reticulata (SNr) is a powerful experimental approach for remote control of different partial seizure types, when targeting the seizure focus is not amenable. Here, we tested the hypothesis that grafting of embryonic/fetal neural precursor cells (NPCs) from various species (rat, human, pig) into STN or SNr of adult rats induces anticonvulsant effects. To rationally refine this approach, we included NPCs derived from the medial ganglionic eminence (MGE) and ventral mesencephalon (VM), both of which are able to develop a GABAergic phenotype. All VM- and MGE-derived cells showed intense migration behavior after grafting into adult rats, developed characteristics of inhibitory interneurons, and survived at least up to 4 months after transplantation. By using the intravenous pentylenetetrazole (PTZ) seizure threshold test in adult rats, transient anticonvulsant effects were observed after bilateral grafting of NPCs derived from human and porcine VM into STN, but not after SNr injection (site-specificity). In contrast, MGE-derived NPCs did not cause anticonvulsant effects after grafting into STN or SNr (cell-specificity). Neither induction of status epilepticus by lithium-pilocarpine to induce neuronal damage prior to the PTZ test nor pretreatment of MGE cells with retinoic acid and potassium chloride to increase differentiation into GABAergic neurons could enhance anticonvulsant effectiveness of MGE cells. This is the first proof-of-principle study showing anticonvulsant effects by bilateral xenotransplantation of NPCs into the STN. Our study highlights the value of VM-derived NPCs for interneuron-based cell grafting targeting the STN.
Collapse
Affiliation(s)
- Bianca Backofen-Wehrhahn
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine Hannover, Germany
| | - Laura Gey
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine Hannover, Germany; Center for Systems Neuroscience, Hannover, Germany
| | - Sonja Bröer
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine Hannover, Germany; Center for Systems Neuroscience, Hannover, Germany
| | - Björn Petersen
- Institute of Farm Animal Genetics, Friedrich-Loeffler-Institut, Mariensee, Germany
| | - Miriam Schiff
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine Hannover, Germany
| | - Annelie Handreck
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine Hannover, Germany; Center for Systems Neuroscience, Hannover, Germany
| | | | - Jessica Scharrenbroich
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine Hannover, Germany
| | - Michael Weißing
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine Hannover, Germany
| | - Selma Staege
- Center for Systems Neuroscience, Hannover, Germany; Department of Neurology, Hannover Medical School, Hannover, Germany
| | - Florian Wegner
- Center for Systems Neuroscience, Hannover, Germany; Department of Neurology, Hannover Medical School, Hannover, Germany
| | - Heiner Niemann
- Institute of Farm Animal Genetics, Friedrich-Loeffler-Institut, Mariensee, Germany
| | - Wolfgang Löscher
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine Hannover, Germany; Center for Systems Neuroscience, Hannover, Germany
| | - Manuela Gernert
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine Hannover, Germany; Center for Systems Neuroscience, Hannover, Germany.
| |
Collapse
|
21
|
Christenson Wick Z, Krook-Magnuson E. Specificity, Versatility, and Continual Development: The Power of Optogenetics for Epilepsy Research. Front Cell Neurosci 2018; 12:151. [PMID: 29962936 PMCID: PMC6010559 DOI: 10.3389/fncel.2018.00151] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2018] [Accepted: 05/15/2018] [Indexed: 12/19/2022] Open
Abstract
Optogenetics is a powerful and rapidly expanding set of techniques that use genetically encoded light sensitive proteins such as opsins. Through the selective expression of these exogenous light-sensitive proteins, researchers gain the ability to modulate neuronal activity, intracellular signaling pathways, or gene expression with spatial, directional, temporal, and cell-type specificity. Optogenetics provides a versatile toolbox and has significantly advanced a variety of neuroscience fields. In this review, using recent epilepsy research as a focal point, we highlight how the specificity, versatility, and continual development of new optogenetic related tools advances our understanding of neuronal circuits and neurological disorders. We additionally provide a brief overview of some currently available optogenetic tools including for the selective expression of opsins.
Collapse
Affiliation(s)
- Zoé Christenson Wick
- Graduate Program in Neuroscience and Department of Neuroscience, University of Minnesota, Minneapolis, MN, United States
| | | |
Collapse
|
22
|
Fink JJ, Levine ES. Uncovering True Cellular Phenotypes: Using Induced Pluripotent Stem Cell-Derived Neurons to Study Early Insults in Neurodevelopmental Disorders. Front Neurol 2018; 9:237. [PMID: 29713304 PMCID: PMC5911479 DOI: 10.3389/fneur.2018.00237] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2017] [Accepted: 03/26/2018] [Indexed: 01/07/2023] Open
Abstract
Animal models of neurodevelopmental disorders have provided invaluable insights into the molecular-, cellular-, and circuit-level defects associated with a plethora of genetic disruptions. In many cases, these deficits have been linked to changes in disease-relevant behaviors, but very few of these findings have been translated to treatments for human disease. This may be due to significant species differences and the difficulty in modeling disorders that involve deletion or duplication of multiple genes. The identification of primary underlying pathophysiology in these models is confounded by the accumulation of secondary disease phenotypes in the mature nervous system, as well as potential compensatory mechanisms. The discovery of induced pluripotent stem cell technology now provides a tool to accurately model complex genetic neurogenetic disorders. Using this technique, patient-specific cell lines can be generated and differentiated into specific subtypes of neurons that can be used to identify primary cellular and molecular phenotypes. It is clear that impairments in synaptic structure and function are a common pathophysiology across neurodevelopmental disorders, and electrophysiological analysis at the earliest stages of neuronal development is critical for identifying changes in activity and excitability that can contribute to synaptic dysfunction and identify targets for disease-modifying therapies.
Collapse
Affiliation(s)
- James J Fink
- Department of Neuroscience, University of Connecticut School of Medicine, Farmington, CT, United States
| | - Eric S Levine
- Department of Neuroscience, University of Connecticut School of Medicine, Farmington, CT, United States
| |
Collapse
|
23
|
Kopach O, Rybachuk O, Krotov V, Kyryk V, Voitenko N, Pivneva T. Maturation of neural stem cells and integration into hippocampal circuits - a functional study in an in situ model of cerebral ischemia. J Cell Sci 2018; 131:jcs.210989. [PMID: 29361548 DOI: 10.1242/jcs.210989] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2017] [Accepted: 12/29/2017] [Indexed: 12/16/2022] Open
Abstract
The hippocampus is the region of the brain that is most susceptible to ischemic lesion because it contains pyramidal neurons that are highly vulnerable to ischemic cell death. A restricted brain neurogenesis limits the possibility of reversing massive cell death after stroke and, hence, endorses cell-based therapies for neuronal replacement strategies following cerebral ischemia. Neurons differentiated from neural stem/progenitor cells (NSPCs) can mature and integrate into host circuitry, improving recovery after stroke. However, how the host environment regulates the NSPC behavior in post-ischemic tissue remains unknown. Here, we studied functional maturation of NSPCs in control and post-ischemic hippocampal tissue after modelling cerebral ischemia in situ We traced the maturation of electrophysiological properties and integration of the NSPC-derived neurons into the host circuits, with these cells developing appropriate activity 3 weeks or less after engraftment. In the tissue subjected to ischemia, the NSPC-derived neurons exhibited functional deficits, and differentiation of embryonic NSPCs to glial types - oligodendrocytes and astrocytes - was boosted. Our findings of the delayed neuronal maturation in post-ischemic conditions, while the NSPC differentiation was promoted towards glial cell types, provide new insights that could be applicable to stem cell therapy replacement strategies used after cerebral ischemia.
Collapse
Affiliation(s)
- Olga Kopach
- Department of Sensory Signalling, Bogomoletz Institute of Physiology, Kyiv 01024, Ukraine .,Institute of Neurology, University College London, London WC1N 3BG, UK
| | - Oksana Rybachuk
- Department of Sensory Signalling, Bogomoletz Institute of Physiology, Kyiv 01024, Ukraine.,State Institute of Genetic and Regenerative Medicine, Kyiv 04114, Ukraine
| | - Volodymyr Krotov
- Department of Sensory Signalling, Bogomoletz Institute of Physiology, Kyiv 01024, Ukraine
| | - Vitalii Kyryk
- State Institute of Genetic and Regenerative Medicine, Kyiv 04114, Ukraine
| | - Nana Voitenko
- Department of Sensory Signalling, Bogomoletz Institute of Physiology, Kyiv 01024, Ukraine.,Kyiv Academic University, Kyiv 03142, Ukraine
| | - Tatyana Pivneva
- Department of Sensory Signalling, Bogomoletz Institute of Physiology, Kyiv 01024, Ukraine.,State Institute of Genetic and Regenerative Medicine, Kyiv 04114, Ukraine.,Kyiv Academic University, Kyiv 03142, Ukraine
| |
Collapse
|
24
|
Terrigno M, Busti I, Alia C, Pietrasanta M, Arisi I, D'Onofrio M, Caleo M, Cremisi F. Neurons Generated by Mouse ESCs with Hippocampal or Cortical Identity Display Distinct Projection Patterns When Co-transplanted in the Adult Brain. Stem Cell Reports 2018; 10:1016-1029. [PMID: 29456186 PMCID: PMC5918192 DOI: 10.1016/j.stemcr.2018.01.010] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2017] [Revised: 01/14/2018] [Accepted: 01/15/2018] [Indexed: 12/14/2022] Open
Abstract
The capability of generating neural precursor cells with distinct types of regional identity in vitro has recently opened new opportunities for cell replacement in animal models of neurodegenerative diseases. By manipulating Wnt and BMP signaling, we steered the differentiation of mouse embryonic stem cells (ESCs) toward isocortical or hippocampal molecular identity. These two types of cells showed different degrees of axonal outgrowth and targeted different regions when co-transplanted in healthy or lesioned isocortex or in hippocampus. In hippocampus, only precursor cells with hippocampal molecular identity were able to extend projections, contacting CA3. Conversely, isocortical-like cells were capable of extending long-range axonal projections only when transplanted in motor cortex, sending fibers toward both intra- and extra-cortical targets. Ischemic damage induced by photothrombosis greatly enhanced the capability of isocortical-like cells to extend far-reaching projections. Our results indicate that neural precursors generated by ESCs carry intrinsic signals specifying axonal extension in different environments.
Wnt signaling induces hippocampal fate in neuralized mouse ESCs Transplanted cortical and hippocampal neurons target distinct regions in adult brain Photothrombotic lesion favors neurite elongation of cortical transplanted cells Cortical cell transplantation improves the motor performance after ischemic damage
Collapse
Affiliation(s)
| | - Irene Busti
- Neurofarba, University of Florence, Florence 50134, Italy; Istituto di Neuroscienze, CNR, Pisa 56124, Italy
| | - Claudia Alia
- Istituto di Neuroscienze, CNR, Pisa 56124, Italy
| | | | - Ivan Arisi
- European Brain Research Institute (EBRI) "Rita Levi-Montalcini", Roma 00161, Italy
| | - Mara D'Onofrio
- European Brain Research Institute (EBRI) "Rita Levi-Montalcini", Roma 00161, Italy
| | - Matteo Caleo
- Istituto di Neuroscienze, CNR, Pisa 56124, Italy
| | | |
Collapse
|
25
|
Directly Converted Human Fibroblasts Mature to Neurons and Show Long-Term Survival in Adult Rodent Hippocampus. Stem Cells Int 2018; 2017:5718608. [PMID: 29317869 PMCID: PMC5727793 DOI: 10.1155/2017/5718608] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2017] [Revised: 10/16/2017] [Accepted: 10/17/2017] [Indexed: 01/21/2023] Open
Abstract
Direct conversion of human somatic cells to induced neurons (iNs), using lineage-specific transcription factors has opened new opportunities for cell therapy in a number of neurological diseases, including epilepsy. In most severe cases of epilepsy, seizures often originate in the hippocampus, where populations of inhibitory interneurons degenerate. Thus, iNs could be of potential use to replace these lost interneurons. It is not known, however, if iNs survive and maintain functional neuronal properties for prolonged time periods in in vivo. We transplanted human fibroblast-derived iNs into the adult rat hippocampus and observed a progressive morphological differentiation, with more developed dendritic arborisation at six months as compared to one month. This was accompanied by mature electrophysiological properties and fast high amplitude action potentials at six months after transplantation. This proof-of-principle study suggests that human iNs can be developed as a candidate source for cell replacement therapy in temporal lobe epilepsy.
Collapse
|
26
|
Single-cell analysis of diversity in human stem cell-derived neurons. Cell Tissue Res 2017; 371:171-179. [PMID: 29185070 DOI: 10.1007/s00441-017-2728-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2017] [Accepted: 11/02/2017] [Indexed: 01/12/2023]
Abstract
Neural stem and progenitor cells produce one of the most remarkable organs in nature, the human brain. Among neural stem cell progeny, post-mitotic neurons are likewise remarkably diverse. Single-cell transcriptomic approaches are now cataloging a long-sought-after molecular taxonomy of neuronal diversity in the brain. Contemporary single-cell omic classifications of neuronal diversity build from electrophysiological approaches that for decades have measured and cataloged diverse biophysical properties of single neurons. With the widespread application of human pluripotent stem cell-based models of neurogenesis to investigate disease pathology and to develop new drugs, a high-resolution understanding of neuronal diversity in vivo is essential to benchmark the state of in vitro models of human neurological disease.
Collapse
|
27
|
Sánchez-Ramón S, Faure F. The Thymus/Neocortex Hypothesis of the Brain: A Cell Basis for Recognition and Instruction of Self. Front Cell Neurosci 2017; 11:340. [PMID: 29163052 PMCID: PMC5663735 DOI: 10.3389/fncel.2017.00340] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2017] [Accepted: 10/13/2017] [Indexed: 12/18/2022] Open
Abstract
The recognition of internal and external sources of stimuli, the self from non-self, seems to be an intrinsic property to the adequate functioning of the immune system and the nervous system, both complex network systems that have evolved to safeguard the self biological identity of the organism. The mammalian brain development relies on dynamic and adaptive processes that are now well described. However, the rules dictating this highly constrained developmental process remain elusive. Here we hypothesize that there is a cellular basis for brain selfhood, based on the analogy of the global mechanisms that drive the self/non-self recognition and instruction by the immune system. In utero education within the thymus by multi-step selection processes discard overly low and high affinity T-lymphocytes to self stimuli, thus avoiding expendable or autoreactive responses that might lead to harmful autoimmunity. We argue that the self principle is one of the chief determinants of neocortical brain neurogenesis. According to our hypothesis, early-life education on self at the subcortical plate of the neocortex by selection processes might participate in the striking specificity of neuronal repertoire and assure efficiency and self tolerance. Potential implications of this hypothesis in self-reactive neurological pathologies are discussed, particularly involving consciousness-associated pathophysiological conditions, i.e., epilepsy and schizophrenia, for which we coined the term autophrenity.
Collapse
Affiliation(s)
- Silvia Sánchez-Ramón
- Department of Clinical Immunology and IdISSC, Hospital Clínico San Carlos, Madrid, Spain.,Department of Microbiology I, School of Medicine, Complutense University of Madrid, Madrid, Spain
| | - Florence Faure
- PSL Research University, INSERM U932, Institut Curie, Paris, France
| |
Collapse
|
28
|
A Look into Stem Cell Therapy: Exploring the Options for Treatment of Ischemic Stroke. Stem Cells Int 2017; 2017:3267352. [PMID: 29201059 PMCID: PMC5671750 DOI: 10.1155/2017/3267352] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2017] [Revised: 08/21/2017] [Accepted: 09/12/2017] [Indexed: 12/14/2022] Open
Abstract
Neural stem cells (NSCs) offer a potential therapeutic benefit in the recovery from ischemic stroke. Understanding the role of endogenous neural stem and progenitor cells under normal physiological conditions aids in analyzing their effects after ischemic injury, including their impact on functional recovery and neurogenesis at the site of injury. Recent animal studies have utilized unique subsets of exogenous and endogenous stem cells as well as preconditioning with pharmacologic agents to better understand the best situation for stem cell proliferation, migration, and differentiation. These stem cell therapies provide a promising effect on stimulation of endogenous neurogenesis, neuroprotection, anti-inflammatory effects, and improved cell survival rates. Clinical trials performed using various stem cell types show promising results to their safety and effectiveness on reducing the effects of ischemic stroke in humans. Another important aspect of stem cell therapy discussed in this review is tracking endogenous and exogenous NSCs with magnetic resonance imaging. This review explores the pathophysiology of NSCs on ischemic stroke, stem cell therapy studies and their effects on neurogenesis, the most recent clinical trials, and techniques to track and monitor the progress of endogenous and exogenous stem cells.
Collapse
|
29
|
Forsberg D, Thonabulsombat C, Jäderstad J, Jäderstad LM, Olivius P, Herlenius E. Functional Stem Cell Integration into Neural Networks Assessed by Organotypic Slice Cultures. ACTA ACUST UNITED AC 2017; 42:2D.13.1-2D.13.30. [PMID: 28806855 DOI: 10.1002/cpsc.34] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Re-formation or preservation of functional, electrically active neural networks has been proffered as one of the goals of stem cell-mediated neural therapeutics. A primary issue for a cell therapy approach is the formation of functional contacts between the implanted cells and the host tissue. Therefore, it is of fundamental interest to establish protocols that allow us to delineate a detailed time course of grafted stem cell survival, migration, differentiation, integration, and functional interaction with the host. One option for in vitro studies is to examine the integration of exogenous stem cells into an existing active neural network in ex vivo organotypic cultures. Organotypic cultures leave the structural integrity essentially intact while still allowing the microenvironment to be carefully controlled. This allows detailed studies over time of cellular responses and cell-cell interactions, which are not readily performed in vivo. This unit describes procedures for using organotypic slice cultures as ex vivo model systems for studying neural stem cell and embryonic stem cell engraftment and communication with CNS host tissue. © 2017 by John Wiley & Sons, Inc.
Collapse
Affiliation(s)
- David Forsberg
- Department of Women's and Children's Health, Karolinska Institutet, and Karolinska University Hospital, Stockholm, Sweden
| | - Charoensri Thonabulsombat
- Department of Clinical Sciences, Intervention and Technology (CLINTEC), Section of Otorhinolaryngology, Karolinska Institutet, and Karolinska University Hospital, Stockholm, Sweden.,Center for Hearing and Communication Research, Karolinska Institutet, and Karolinska University Hospital, Stockholm, Sweden.,Department of Anatomy, Faculty of Science, Mahidol University, Bangkok, Thailand
| | - Johan Jäderstad
- Department of Women's and Children's Health, Karolinska Institutet, and Karolinska University Hospital, Stockholm, Sweden
| | - Linda Maria Jäderstad
- Department of Women's and Children's Health, Karolinska Institutet, and Karolinska University Hospital, Stockholm, Sweden
| | - Petri Olivius
- Department of Clinical Sciences, Intervention and Technology (CLINTEC), Section of Otorhinolaryngology, Karolinska Institutet, and Karolinska University Hospital, Stockholm, Sweden.,Center for Hearing and Communication Research, Karolinska Institutet, and Karolinska University Hospital, Stockholm, Sweden
| | - Eric Herlenius
- Department of Women's and Children's Health, Karolinska Institutet, and Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
30
|
Shimamura N, Katagai T, Kakuta K, Matsuda N, Katayama K, Fujiwara N, Watanabe Y, Naraoka M, Ohkuma H. Rehabilitation and the Neural Network After Stroke. Transl Stroke Res 2017; 8:507-514. [PMID: 28681346 DOI: 10.1007/s12975-017-0550-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2017] [Revised: 06/23/2017] [Accepted: 06/26/2017] [Indexed: 12/18/2022]
Abstract
Stroke remains a major cause of disability throughout the world: paralysis, cognitive impairment, aphasia, and so on. Surgical or medical intervention is curative in only a small number of cases. Nearly all stroke cases require rehabilitation. Neurorehabilitation generally improves patient outcome, but it sometimes has no effect or even a mal-influence. The aim of this review is the clarification of the mechanisms of neurorehabilitation. We systematically reviewed recently published articles on neural network remodeling, especially from 2014 to 2016. Finally, we summarize progress in neurorehabilitation and discuss future prospects.
Collapse
Affiliation(s)
- Norihito Shimamura
- Department of Neurosurgery, Hirosaki University School of Medicine, 5-Zaihuchou, Hirosaki, Aomori, 036-8562, Japan.
| | - Takeshi Katagai
- Department of Neurosurgery, Hirosaki University School of Medicine, 5-Zaihuchou, Hirosaki, Aomori, 036-8562, Japan
| | - Kiyohide Kakuta
- Department of Neurosurgery, Hirosaki University School of Medicine, 5-Zaihuchou, Hirosaki, Aomori, 036-8562, Japan
| | - Naoya Matsuda
- Department of Neurosurgery, Hirosaki University School of Medicine, 5-Zaihuchou, Hirosaki, Aomori, 036-8562, Japan
| | - Kosuke Katayama
- Department of Neurosurgery, Hirosaki University School of Medicine, 5-Zaihuchou, Hirosaki, Aomori, 036-8562, Japan
| | - Nozomi Fujiwara
- Department of Neurosurgery, Hirosaki University School of Medicine, 5-Zaihuchou, Hirosaki, Aomori, 036-8562, Japan
| | - Yuuka Watanabe
- Department of Neurosurgery, Hirosaki University School of Medicine, 5-Zaihuchou, Hirosaki, Aomori, 036-8562, Japan
| | - Masato Naraoka
- Department of Neurosurgery, Hirosaki University School of Medicine, 5-Zaihuchou, Hirosaki, Aomori, 036-8562, Japan
| | - Hiroki Ohkuma
- Department of Neurosurgery, Hirosaki University School of Medicine, 5-Zaihuchou, Hirosaki, Aomori, 036-8562, Japan
| |
Collapse
|
31
|
Tornero D, Tsupykov O, Granmo M, Rodriguez C, Grønning-Hansen M, Thelin J, Smozhanik E, Laterza C, Wattananit S, Ge R, Tatarishvili J, Grealish S, Brüstle O, Skibo G, Parmar M, Schouenborg J, Lindvall O, Kokaia Z. Synaptic inputs from stroke-injured brain to grafted human stem cell-derived neurons activated by sensory stimuli. Brain 2017; 140:692-706. [PMID: 28115364 DOI: 10.1093/brain/aww347] [Citation(s) in RCA: 70] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2016] [Accepted: 11/20/2016] [Indexed: 12/20/2022] Open
Abstract
Transplanted neurons derived from stem cells have been proposed to improve function in animal models of human disease by various mechanisms such as neuronal replacement. However, whether the grafted neurons receive functional synaptic inputs from the recipient's brain and integrate into host neural circuitry is unknown. Here we studied the synaptic inputs from the host brain to grafted cortical neurons derived from human induced pluripotent stem cells after transplantation into stroke-injured rat cerebral cortex. Using the rabies virus-based trans-synaptic tracing method and immunoelectron microscopy, we demonstrate that the grafted neurons receive direct synaptic inputs from neurons in different host brain areas located in a pattern similar to that of neurons projecting to the corresponding endogenous cortical neurons in the intact brain. Electrophysiological in vivo recordings from the cortical implants show that physiological sensory stimuli, i.e. cutaneous stimulation of nose and paw, can activate or inhibit spontaneous activity in grafted neurons, indicating that at least some of the afferent inputs are functional. In agreement, we find using patch-clamp recordings that a portion of grafted neurons respond to photostimulation of virally transfected, channelrhodopsin-2-expressing thalamo-cortical axons in acute brain slices. The present study demonstrates, for the first time, that the host brain regulates the activity of grafted neurons, providing strong evidence that transplanted human induced pluripotent stem cell-derived cortical neurons can become incorporated into injured cortical circuitry. Our findings support the idea that these neurons could contribute to functional recovery in stroke and other conditions causing neuronal loss in cerebral cortex.
Collapse
Affiliation(s)
- Daniel Tornero
- Laboratory of Stem Cells and Restorative Neurology, Lund Stem Cell Center, University Hospital, BMC B10, 221 84, Lund, Sweden
| | - Oleg Tsupykov
- Bogomoletz Institute of Physiology, and State Institute of Genetic and Regenerative Medicine, 01024, Kyiv, Ukraine
| | - Marcus Granmo
- Neuronano Research Center, Lund University, Scheelevägen 2, 223 81, Lund, Sweden
| | - Cristina Rodriguez
- Laboratory of Stem Cells and Restorative Neurology, Lund Stem Cell Center, University Hospital, BMC B10, 221 84, Lund, Sweden
| | - Marita Grønning-Hansen
- Laboratory of Stem Cells and Restorative Neurology, Lund Stem Cell Center, University Hospital, BMC B10, 221 84, Lund, Sweden
| | - Jonas Thelin
- Neuronano Research Center, Lund University, Scheelevägen 2, 223 81, Lund, Sweden
| | - Ekaterina Smozhanik
- Bogomoletz Institute of Physiology, and State Institute of Genetic and Regenerative Medicine, 01024, Kyiv, Ukraine
| | - Cecilia Laterza
- Laboratory of Stem Cells and Restorative Neurology, Lund Stem Cell Center, University Hospital, BMC B10, 221 84, Lund, Sweden
| | - Somsak Wattananit
- Laboratory of Stem Cells and Restorative Neurology, Lund Stem Cell Center, University Hospital, BMC B10, 221 84, Lund, Sweden
| | - Ruimin Ge
- Laboratory of Stem Cells and Restorative Neurology, Lund Stem Cell Center, University Hospital, BMC B10, 221 84, Lund, Sweden
| | - Jemal Tatarishvili
- Laboratory of Stem Cells and Restorative Neurology, Lund Stem Cell Center, University Hospital, BMC B10, 221 84, Lund, Sweden
| | - Shane Grealish
- Developmental and Regenerative Neurobiology, Department of Experimental Medical Science, Wallenberg Neuroscience Center and Lund Stem Cell Center, Lund University, BMC A11, 221 84, Lund, Sweden
| | - Oliver Brüstle
- Institute of Reconstructive Neurobiology, Life and Brain Center, University of Bonn, and German Center for Neurodegenerative Diseases (DZNE), Sigmund-Freud-Straße 25, D-53127, Bonn, Germany
| | - Galina Skibo
- Bogomoletz Institute of Physiology, and State Institute of Genetic and Regenerative Medicine, 01024, Kyiv, Ukraine
| | - Malin Parmar
- Developmental and Regenerative Neurobiology, Department of Experimental Medical Science, Wallenberg Neuroscience Center and Lund Stem Cell Center, Lund University, BMC A11, 221 84, Lund, Sweden
| | - Jens Schouenborg
- Neuronano Research Center, Lund University, Scheelevägen 2, 223 81, Lund, Sweden
| | - Olle Lindvall
- Laboratory of Stem Cells and Restorative Neurology, Lund Stem Cell Center, University Hospital, BMC B10, 221 84, Lund, Sweden
| | - Zaal Kokaia
- Laboratory of Stem Cells and Restorative Neurology, Lund Stem Cell Center, University Hospital, BMC B10, 221 84, Lund, Sweden
| |
Collapse
|
32
|
Niclis JC, Turner C, Durnall J, McDougal S, Kauhausen JA, Leaw B, Dottori M, Parish CL, Thompson LH. Long-Distance Axonal Growth and Protracted Functional Maturation of Neurons Derived from Human Induced Pluripotent Stem Cells After Intracerebral Transplantation. Stem Cells Transl Med 2017; 6:1547-1556. [PMID: 28198124 PMCID: PMC5689777 DOI: 10.1002/sctm.16-0198] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2016] [Accepted: 10/31/2016] [Indexed: 12/21/2022] Open
Abstract
The capacity for induced pluripotent stem (iPS) cells to be differentiated into a wide range of neural cell types makes them an attractive donor source for autologous neural transplantation therapies aimed at brain repair. Translation to the in vivo setting has been difficult, however, with mixed results in a wide variety of preclinical models of brain injury and limited information on the basic in vivo properties of neural grafts generated from human iPS cells. Here we have generated a human iPS cell line constitutively expressing green fluorescent protein as a basis to identify and characterize grafts resulting from transplantation of neural progenitors into the adult rat brain. The results show that the grafts contain a mix of neural cell types, at various stages of differentiation, including neurons that establish extensive patterns of axonal growth and progressively develop functional properties over the course of 1 year after implantation. These findings form an important basis for the design and interpretation of preclinical studies using human stem cells for functional circuit re‐construction in animal models of brain injury. Stem Cells Translational Medicine2017;6:1547–1556
Collapse
Affiliation(s)
- Jonathan C Niclis
- Florey Institute for Neuroscience and Mental Health, Royal Parade, Parkville, Victoria, Australia
| | - Christopher Turner
- Florey Institute for Neuroscience and Mental Health, Royal Parade, Parkville, Victoria, Australia
| | - Jennifer Durnall
- Florey Institute for Neuroscience and Mental Health, Royal Parade, Parkville, Victoria, Australia
| | - Stuart McDougal
- Florey Institute for Neuroscience and Mental Health, Royal Parade, Parkville, Victoria, Australia
| | - Jessica A Kauhausen
- Florey Institute for Neuroscience and Mental Health, Royal Parade, Parkville, Victoria, Australia
| | - Bryan Leaw
- Florey Institute for Neuroscience and Mental Health, Royal Parade, Parkville, Victoria, Australia
| | - Mirella Dottori
- Department of Electrical and Electronic Engineering, Centre for Neural Engineering, University of Melbourne, Royal Parade, Parkville, Victoria, Australia
| | - Clare L Parish
- Florey Institute for Neuroscience and Mental Health, Royal Parade, Parkville, Victoria, Australia
| | - Lachlan H Thompson
- Florey Institute for Neuroscience and Mental Health, Royal Parade, Parkville, Victoria, Australia
| |
Collapse
|
33
|
Martínez-Serrano A, Pereira MP, Avaliani N, Nelke A, Kokaia M, Ramos-Moreno T. Short-Term Grafting of Human Neural Stem Cells: Electrophysiological Properties and Motor Behavioral Amelioration in Experimental Parkinson's Disease. Cell Transplant 2016; 25:2083-2097. [DOI: 10.3727/096368916x692069] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Cell replacement therapy in Parkinson's disease (PD) still lacks a study addressing the acquisition of electrophysiological properties of human grafted neural stem cells and their relation with the emergence of behavioral recovery after transplantation in the short term. Here we study the electrophysiological and biochemical profiles of two ventral mesencephalic human neural stem cell (NSC) clonal lines (C30-Bcl-XL and C32-Bcl-XL) that express high levels of Bcl-XL to enhance their neurogenic capacity, after grafting in an in vitro parkinsonian model. Electrophysiological recordings show that the majority of the cells derived from the transplants are not mature at 6 weeks after grafting, but 6.7% of the studied cells showed mature electrophysiological profiles. Nevertheless, parallel in vivo behavioral studies showed a significant motor improvement at 7 weeks postgrafting in the animals receiving C30-Bcl-XL, the cell line producing the highest amount of TH+ cells. Present results show that, at this postgrafting time point, behavioral amelioration highly correlates with the spatial dispersion of the TH+ grafted cells in the caudate putamen. The spatial dispersion, along with a high number of dopaminergic-derived cells, is crucial for behavioral improvements. Our findings have implications for long-term standardization of stem cell-based approaches in Parkinson's disease.
Collapse
Affiliation(s)
- Alberto Martínez-Serrano
- Department of Molecular Biology, Universidad Autónoma de Madrid, Madrid, Spain
- Department of Molecular Neurobiology, Center of Molecular Biology Severo Ochoa (UAM-CSIC), Madrid, Spain
| | - Marta P. Pereira
- Department of Molecular Biology, Universidad Autónoma de Madrid, Madrid, Spain
- Department of Molecular Neurobiology, Center of Molecular Biology Severo Ochoa (UAM-CSIC), Madrid, Spain
| | - Natalia Avaliani
- Epilepsy Center/Stem Cell Center, Wallenberg Neuroscience Center, Lund University Hospital, Lund, Sweden
| | - Anna Nelke
- Department of Molecular Biology, Universidad Autónoma de Madrid, Madrid, Spain
- Department of Molecular Neurobiology, Center of Molecular Biology Severo Ochoa (UAM-CSIC), Madrid, Spain
| | - Merab Kokaia
- Epilepsy Center/Stem Cell Center, Wallenberg Neuroscience Center, Lund University Hospital, Lund, Sweden
| | - Tania Ramos-Moreno
- Department of Molecular Biology, Universidad Autónoma de Madrid, Madrid, Spain
- Department of Molecular Neurobiology, Center of Molecular Biology Severo Ochoa (UAM-CSIC), Madrid, Spain
- Epilepsy Center/Stem Cell Center, Wallenberg Neuroscience Center, Lund University Hospital, Lund, Sweden
| |
Collapse
|
34
|
Avaliani N, Andersson M, Runegaard AH, Woldbye D, Kokaia M. DREADDs suppress seizure-like activity in a mouse model of pharmacoresistant epileptic brain tissue. Gene Ther 2016; 23:760-766. [PMID: 27416078 DOI: 10.1038/gt.2016.56] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2016] [Accepted: 06/20/2016] [Indexed: 12/11/2022]
Abstract
Epilepsy is a neurological disorder with a prevalence of ≈1% of general population. Available antiepileptic drugs (AEDs) have multiple side effects and are ineffective in 30% of patients. Therefore, development of effective treatment strategies is highly needed, requiring drug-screening models that are relevant and reliable. We investigated novel chemogenetic approach, using DREADDs (designer receptors exclusively activated by designer drugs) as possible inhibitor of epileptiform activity in organotypic hippocampal slice cultures (OHSCs). The OHSCs are characterized by increased overall excitability and closely resemble features of human epileptic tissue. Studies suggest that chemically induced epileptiform activity in rat OHSCs is pharmacoresistant to most of AEDs. However, high-frequency electric stimulus train-induced bursting (STIB) in OHSCs is responsive to carbamazepine and phenytoin. We investigated whether inhibitory DREADD, hM4Di, would be effective in suppressing STIB in OHSC. hM4Di is a mutated muscarinic receptor selectively activated by otherwise inert clozapine-N-oxide, which leads to hyperpolarization in neurons. We demonstrated that this hyperpolarization effectively suppresses STIB in mouse OHSCs. As we also found that STIB in mouse OHSCs is resistant to common AED, valproic acid, collectively our findings suggest that DREADD-based strategy may be effective in suppressing epileptiform activity in a pharamcoresitant epileptic brain tissue.
Collapse
Affiliation(s)
- N Avaliani
- Epilepsy Centre, Experimental Epilepsy Group, Department of Clinical Sciences, Lund University Hospital, Lund, Sweden
| | - M Andersson
- Epilepsy Centre, Experimental Epilepsy Group, Department of Clinical Sciences, Lund University Hospital, Lund, Sweden
| | - A H Runegaard
- Molecular Neuropharmacology and Genetics Laboratory, Department of Neuroscience and Pharmacology, University of Copenhagen, Copenhagen, Denmark
| | - D Woldbye
- Laboratory of Neural Plasticity, Department of Neuroscience and Pharmacology, University of Copenhagen, Copenhagen, Denmark
| | - M Kokaia
- Epilepsy Centre, Experimental Epilepsy Group, Department of Clinical Sciences, Lund University Hospital, Lund, Sweden
| |
Collapse
|
35
|
Bui AD, Alexander A, Soltesz I. Seizing Control: From Current Treatments to Optogenetic Interventions in Epilepsy. Neuroscientist 2016; 23:68-81. [PMID: 26700888 DOI: 10.1177/1073858415619600] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
The unpredictability and severity of seizures contribute to the debilitating nature of epilepsy. These factors also render the condition particularly challenging to treat, as an ideal treatment would need to detect and halt the pathological bursts of hyperactivity without disrupting normal brain activity. Optogenetic techniques offer promising tools to study and perhaps eventually treat this episodic disorder by controlling specific brain circuits in epileptic animals with great temporal precision. Here, we briefly review the current treatment options for patients with epilepsy. We then describe the many ways optogenetics has allowed us to untangle the microcircuits involved in seizure activity, and how it has, in some cases, changed our perception of previous theories of seizure generation. Control of seizures with light is no longer a dream, and has been achieved in numerous different animal models of epilepsy. Beyond its application as a seizure suppressor, we highlight another facet of optogenetics in epilepsy, namely the ability to create "on-demand" seizures, as a tool to systematically probe the dynamics of networks during seizure initiation and propagation. Finally, we look into the future to discuss the possibilities and challenges of translating optogenetic techniques to clinical use.
Collapse
Affiliation(s)
- Anh D Bui
- 1 Department of Neurosurgery, Stanford University, Stanford, CA, USA.,2 Department of Anatomy and Neurobiology, University of California, Irvine, CA, USA
| | - Allyson Alexander
- 1 Department of Neurosurgery, Stanford University, Stanford, CA, USA
| | - Ivan Soltesz
- 1 Department of Neurosurgery, Stanford University, Stanford, CA, USA
| |
Collapse
|
36
|
Optogenetic control of human neurons in organotypic brain cultures. Sci Rep 2016; 6:24818. [PMID: 27098488 PMCID: PMC4838935 DOI: 10.1038/srep24818] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2015] [Accepted: 04/06/2016] [Indexed: 01/16/2023] Open
Abstract
Optogenetics is one of the most powerful tools in neuroscience, allowing for selective control of specific neuronal populations in the brain of experimental animals, including mammals. We report, for the first time, the application of optogenetic tools to human brain tissue providing a proof-of-concept for the use of optogenetics in neuromodulation of human cortical and hippocampal neurons as a possible tool to explore network mechanisms and develop future therapeutic strategies.
Collapse
|
37
|
Lietman SA. Induced pluripotent stem cells in cartilage repair. World J Orthop 2016; 7:149-155. [PMID: 27004161 PMCID: PMC4794532 DOI: 10.5312/wjo.v7.i3.149] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2015] [Revised: 11/17/2015] [Accepted: 01/04/2016] [Indexed: 02/06/2023] Open
Abstract
Articular cartilage repair techniques are challenging. Human embryonic stem cells and induced pluripotent stem cells (iPSCs) theoretically provide an unlimited number of specialized cells which could be used in articular cartilage repair. However thus far chondrocytes from iPSCs have been created primarily by viral transfection and with the use of cocultured feeder cells. In addition chondrocytes derived from iPSCs have usually been formed in condensed cell bodies (resembling embryoid bodies) that then require dissolution with consequent substantial loss of cell viability and phenotype. All of these current techniques used to derive chondrocytes from iPSCs are problematic but solutions to these problems are on the horizon. These solutions will make iPSCs a viable alternative for articular cartilage repair in the near future.
Collapse
|
38
|
Shining Light on the Sprout of Life: Optogenetics Applications in Stem Cell Research and Therapy. J Membr Biol 2016; 249:215-20. [DOI: 10.1007/s00232-016-9883-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2015] [Accepted: 02/18/2016] [Indexed: 12/21/2022]
|
39
|
Holmqvist S, Lehtonen Š, Chumarina M, Puttonen KA, Azevedo C, Lebedeva O, Ruponen M, Oksanen M, Djelloul M, Collin A, Goldwurm S, Meyer M, Lagarkova M, Kiselev S, Koistinaho J, Roybon L. Creation of a library of induced pluripotent stem cells from Parkinsonian patients. NPJ Parkinsons Dis 2016; 2:16009. [PMID: 28725696 PMCID: PMC5516589 DOI: 10.1038/npjparkd.2016.9] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2015] [Revised: 04/01/2016] [Accepted: 04/11/2016] [Indexed: 02/07/2023] Open
Abstract
Induced pluripotent stem cells (iPSCs) are becoming an important source of pre-clinical models for research focusing on neurodegeneration. They offer the possibility for better understanding of common and divergent pathogenic mechanisms of brain diseases. Moreover, iPSCs provide a unique opportunity to develop personalized therapeutic strategies, as well as explore early pathogenic mechanisms, since they rely on the use of patients' own cells that are otherwise accessible only post-mortem, when neuronal death-related cellular pathways and processes are advanced and adaptive. Neurodegenerative diseases are in majority of unknown cause, but mutations in specific genes can lead to familial forms of these diseases. For example, mutations in the superoxide dismutase 1 gene lead to the motor neuron disease amyotrophic lateral sclerosis (ALS), while mutations in the SNCA gene encoding for alpha-synuclein protein lead to familial Parkinson's disease (PD). The generations of libraries of familial human ALS iPSC lines have been described, and the iPSCs rapidly became useful models for studying cell autonomous and non-cell autonomous mechanisms of the disease. Here we report the generation of a comprehensive library of iPSC lines of familial PD and an associated synucleinopathy, multiple system atrophy (MSA). In addition, we provide examples of relevant neural cell types these iPSC can be differentiated into, and which could be used to further explore early disease mechanisms. These human cellular models will be a valuable resource for identifying common and divergent mechanisms leading to neurodegeneration in PD and MSA.
Collapse
Affiliation(s)
- Staffan Holmqvist
- Stem Cell Laboratory for CNS Disease Modeling, Wallenberg Neuroscience Center, Department of Experimental Medical Science, BMC A10, Lund University, Lund, Sweden
- Strategic Research Area MultiPark, Lund University, Lund, Sweden
- Lund Stem Cell Center, Lund University, Lund, Sweden
| | - Šárka Lehtonen
- Stem Cell Laboratory of Molecular Brain Research Group, Department of Neurobiology, A.I. Virtanen Institute, University of Eastern Finland, Kuopio, Finland
| | - Margarita Chumarina
- Stem Cell Laboratory for CNS Disease Modeling, Wallenberg Neuroscience Center, Department of Experimental Medical Science, BMC A10, Lund University, Lund, Sweden
- Strategic Research Area MultiPark, Lund University, Lund, Sweden
- Lund Stem Cell Center, Lund University, Lund, Sweden
| | - Katja A Puttonen
- Stem Cell Laboratory of Molecular Brain Research Group, Department of Neurobiology, A.I. Virtanen Institute, University of Eastern Finland, Kuopio, Finland
| | - Carla Azevedo
- Stem Cell Laboratory for CNS Disease Modeling, Wallenberg Neuroscience Center, Department of Experimental Medical Science, BMC A10, Lund University, Lund, Sweden
- Strategic Research Area MultiPark, Lund University, Lund, Sweden
- Lund Stem Cell Center, Lund University, Lund, Sweden
| | - Olga Lebedeva
- Russian Academy of Sciences, Vavilov Institute of General Genetics, Moscow, Russia
| | - Marika Ruponen
- Stem Cell Laboratory of Molecular Brain Research Group, Department of Neurobiology, A.I. Virtanen Institute, University of Eastern Finland, Kuopio, Finland
| | - Minna Oksanen
- Stem Cell Laboratory of Molecular Brain Research Group, Department of Neurobiology, A.I. Virtanen Institute, University of Eastern Finland, Kuopio, Finland
| | - Mehdi Djelloul
- Stem Cell Laboratory for CNS Disease Modeling, Wallenberg Neuroscience Center, Department of Experimental Medical Science, BMC A10, Lund University, Lund, Sweden
- Strategic Research Area MultiPark, Lund University, Lund, Sweden
- Lund Stem Cell Center, Lund University, Lund, Sweden
| | - Anna Collin
- Department of Clinical Genetics and Biobanks, Office for Medical Services, Division of Laboratory Medicine, Lund, Sweden
| | - Stefano Goldwurm
- Parkinson Institute, Istituti Clinici di Perfezionamento, Milan, Italy
| | - Morten Meyer
- Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Maria Lagarkova
- Russian Academy of Sciences, Vavilov Institute of General Genetics, Moscow, Russia
| | - Sergei Kiselev
- Russian Academy of Sciences, Vavilov Institute of General Genetics, Moscow, Russia
| | - Jari Koistinaho
- Stem Cell Laboratory of Molecular Brain Research Group, Department of Neurobiology, A.I. Virtanen Institute, University of Eastern Finland, Kuopio, Finland
- ()
| | - Laurent Roybon
- Stem Cell Laboratory for CNS Disease Modeling, Wallenberg Neuroscience Center, Department of Experimental Medical Science, BMC A10, Lund University, Lund, Sweden
- Strategic Research Area MultiPark, Lund University, Lund, Sweden
- Lund Stem Cell Center, Lund University, Lund, Sweden
- ()
| |
Collapse
|
40
|
Li X, Bao X, Wang R. Experimental models of Alzheimer's disease for deciphering the pathogenesis and therapeutic screening (Review). Int J Mol Med 2015; 37:271-83. [PMID: 26676932 DOI: 10.3892/ijmm.2015.2428] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2015] [Accepted: 12/02/2015] [Indexed: 11/06/2022] Open
Abstract
Despite decades of laboratory and clinical research, Alzheimer's disease (AD) is still the leading cause of dementia in adults and there are no curative therapies currently available for this disease. This may be due to the pathological features of AD, which include extensive extracellular amyloid plaques and intracellular neurofibrillary tangles, as well as subsequent neuronal and synaptic loss, which begin to appear several years prior to memory loss and the damge is already irreversible and extensive at the time of clinical diagnosis. The poor therapeutic effects of current treatments necessitate the introduction of experimental models able to replicate AD pathology, particularly in the pre-symptomatic stage, and then to explore preventive and therapeutic strategies. In response to this necessity, various experimental models reproducing human AD pathology have been developed, which are also useful tools for therapeutic screening. Although none of these models fully reproduce the key features of human AD, the experimental models do provide important insight into the pathological changes which occur in AD. This review summarizes the commonly used experimental models of AD and also discusses how the models may be used to decipher the pathogenesis underlying AD and to screen novel therapies for this disease.
Collapse
Affiliation(s)
- Xueyuan Li
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Dongdan, Dong Cheng, Beijing 100005, P.R. China
| | - Xinjie Bao
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Dongdan, Dong Cheng, Beijing 100005, P.R. China
| | - Renzhi Wang
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Dongdan, Dong Cheng, Beijing 100005, P.R. China
| |
Collapse
|
41
|
Nestor MW, Phillips AW, Artimovich E, Nestor JE, Hussman JP, Blatt GJ. Human Inducible Pluripotent Stem Cells and Autism Spectrum Disorder: Emerging Technologies. Autism Res 2015; 9:513-35. [DOI: 10.1002/aur.1570] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2015] [Revised: 09/03/2015] [Accepted: 09/08/2015] [Indexed: 12/11/2022]
Affiliation(s)
- Michael W. Nestor
- The Hussman Institute for Autism; 801 W. Baltimore St., Suite 301 Baltimore Maryland 21201
| | - Andre W. Phillips
- The Hussman Institute for Autism; 801 W. Baltimore St., Suite 301 Baltimore Maryland 21201
| | - Elena Artimovich
- The Hussman Institute for Autism; 801 W. Baltimore St., Suite 301 Baltimore Maryland 21201
| | - Jonathan E. Nestor
- The Hussman Institute for Autism; 801 W. Baltimore St., Suite 301 Baltimore Maryland 21201
| | - John P. Hussman
- The Hussman Institute for Autism; 801 W. Baltimore St., Suite 301 Baltimore Maryland 21201
| | - Gene J. Blatt
- The Hussman Institute for Autism; 801 W. Baltimore St., Suite 301 Baltimore Maryland 21201
| |
Collapse
|
42
|
Shimba K, Sakai K, Takayama Y, Kotani K, Jimbo Y. Recording axonal conduction to evaluate the integration of pluripotent cell-derived neurons into a neuronal network. Biomed Microdevices 2015; 17:94. [PMID: 26303583 DOI: 10.1007/s10544-015-9997-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Stem cell transplantation is a promising therapy to treat neurodegenerative disorders, and a number of in vitro models have been developed for studying interactions between grafted neurons and the host neuronal network to promote drug discovery. However, methods capable of evaluating the process by which stem cells integrate into the host neuronal network are lacking. In this study, we applied an axonal conduction-based analysis to a co-culture study of primary and differentiated neurons. Mouse cortical neurons and neuronal cells differentiated from P19 embryonal carcinoma cells, a model for early neural differentiation of pluripotent stem cells, were co-cultured in a microfabricated device. The somata of these cells were separated by the co-culture device, but their axons were able to elongate through microtunnels and then form synaptic contacts. Propagating action potentials were recorded from these axons by microelectrodes embedded at the bottom of the microtunnels and sorted into clusters representing individual axons. While the number of axons of cortical neurons increased until 14 days in vitro and then decreased, those of P19 neurons increased throughout the culture period. Network burst analysis showed that P19 neurons participated in approximately 80% of the bursting activity after 14 days in vitro. Interestingly, the axonal conduction delay of P19 neurons was significantly greater than that of cortical neurons, suggesting that there are some physiological differences in their axons. These results suggest that our method is feasible to evaluate the process by which stem cell-derived neurons integrate into a host neuronal network.
Collapse
Affiliation(s)
- Kenta Shimba
- Department of Human and Engineered Environmental Studies, Graduate School of Frontier Sciences, University of Tokyo, Room 1122, Faculty of Engineering Bldg., 14, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8656, Japan,
| | | | | | | | | |
Collapse
|
43
|
Abstract
Organotypic hippocampal slice cultures (OHSCs) have been used as a powerful ex vivo model for decades. They have been used successfully in studies of neuronal death, microglial activation, mossy fiber regeneration, neurogenesis, and drug screening. As a pre-animal experimental phase for physiologic and pathologic brain research, OHSCs offer outcomes that are relatively closer to those of whole-animal studies than outcomes obtained from cell culture in vitro. At the same time, mechanisms can be studied more precisely in OHSCs than they can be in vivo. Here, we summarize stroke and traumatic brain injury research that has been carried out in OHSCs and review classic experimental applications of OHSCs and its limitations.
Collapse
|
44
|
Chinchalongporn V, Koppensteiner P, Prè D, Thangnipon W, Bilo L, Arancio O. Connectivity and circuitry in a dish versus in a brain. ALZHEIMERS RESEARCH & THERAPY 2015; 7:44. [PMID: 26045718 PMCID: PMC4456047 DOI: 10.1186/s13195-015-0129-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
In order to understand and find therapeutic strategies for neurological disorders, disease models that recapitulate the connectivity and circuitry of patients’ brain are needed. Owing to many limitations of animal disease models, in vitro neuronal models using patient-derived stem cells are currently being developed. However, prior to employing neurons as a model in a dish, they need to be evaluated for their electrophysiological properties, including both passive and active membrane properties, dynamics of neurotransmitter release, and capacity to undergo synaptic plasticity. In this review, we survey recent attempts to study these issues in human induced pluripotent stem cell-derived neurons. Although progress has been made, there are still many hurdles to overcome before human induced pluripotent stem cell-derived neurons can fully recapitulate all of the above physiological properties of adult mature neurons. Moreover, proper integration of neurons into pre-existing circuitry still needs to be achieved. Nevertheless, in vitro neuronal stem cell-derived models hold great promise for clinical application in neurological diseases in the future.
Collapse
Affiliation(s)
- Vorapin Chinchalongporn
- Department of Pathology & Cell Biology, Columbia University, New York, NY 10032 USA ; Taub Institute for Research on Alzheimer's Disease and the Aging Brain P&S Bldg, Room 12-420D, Columbia University, New York, NY 10032 USA ; Columbia Stem Cell Initiative, CUMC, New York, NY 10032 USA ; Research Center for Neuroscience, Institute of Molecular Biosciences, Mahidol University, Salaya, Nakhonpathom 73170 Thailand
| | - Peter Koppensteiner
- Department of Pathology & Cell Biology, Columbia University, New York, NY 10032 USA ; Taub Institute for Research on Alzheimer's Disease and the Aging Brain P&S Bldg, Room 12-420D, Columbia University, New York, NY 10032 USA ; Columbia Stem Cell Initiative, CUMC, New York, NY 10032 USA ; Department of Neurophysiology and Neuropharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, 1090 Vienna, Austria
| | - Deborah Prè
- Department of Pathology & Cell Biology, Columbia University, New York, NY 10032 USA ; Taub Institute for Research on Alzheimer's Disease and the Aging Brain P&S Bldg, Room 12-420D, Columbia University, New York, NY 10032 USA ; Columbia Stem Cell Initiative, CUMC, New York, NY 10032 USA
| | - Wipawan Thangnipon
- Research Center for Neuroscience, Institute of Molecular Biosciences, Mahidol University, Salaya, Nakhonpathom 73170 Thailand
| | - Leonilda Bilo
- Department of Pathology & Cell Biology, Columbia University, New York, NY 10032 USA ; Taub Institute for Research on Alzheimer's Disease and the Aging Brain P&S Bldg, Room 12-420D, Columbia University, New York, NY 10032 USA ; Columbia Stem Cell Initiative, CUMC, New York, NY 10032 USA ; Department of Neurosciences, Reproductive and Odontostomatological Sciences, Federico II University of Naples, 80131 Naples, Italy
| | - Ottavio Arancio
- Department of Pathology & Cell Biology, Columbia University, New York, NY 10032 USA ; Taub Institute for Research on Alzheimer's Disease and the Aging Brain P&S Bldg, Room 12-420D, Columbia University, New York, NY 10032 USA ; Columbia Stem Cell Initiative, CUMC, New York, NY 10032 USA
| |
Collapse
|
45
|
Thompson LH, Björklund A. Reconstruction of brain circuitry by neural transplants generated from pluripotent stem cells. Neurobiol Dis 2015; 79:28-40. [PMID: 25913029 DOI: 10.1016/j.nbd.2015.04.003] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2015] [Revised: 04/09/2015] [Accepted: 04/15/2015] [Indexed: 12/15/2022] Open
Abstract
Pluripotent stem cells (embryonic stem cells, ESCs, and induced pluripotent stem cells, iPSCs) have the capacity to generate neural progenitors that are intrinsically patterned to undergo differentiation into specific neuronal subtypes and express in vivo properties that match the ones formed during normal embryonic development. Remarkable progress has been made in this field during recent years thanks to the development of more refined protocols for the generation of transplantable neuronal progenitors from pluripotent stem cells, and the access to new tools for tracing of neuronal connectivity and assessment of integration and function of grafted neurons. Recent studies in brains of neonatal mice or rats, as well as in rodent models of brain or spinal cord damage, have shown that ESC- or iPSC-derived neural progenitors can be made to survive and differentiate after transplantation, and that they possess a remarkable capacity to extend axons over long distances and become functionally integrated into host neural circuitry. Here, we summarize these recent developments in the perspective of earlier studies using intracerebral and intraspinal transplants of primary neurons derived from fetal brain, with special focus on the ability of human ESC- and iPSC-derived progenitors to reconstruct damaged neural circuitry in cortex, hippocampus, the nigrostriatal system and the spinal cord, and we discuss the intrinsic and extrinsic factors that determine the growth properties of the grafted neurons and their capacity to establish target-specific long-distance axonal connections in the damaged host brain.
Collapse
Affiliation(s)
- Lachlan H Thompson
- Florey Institute for Neuroscience and Mental Health, University of Melbourne, Royal Parade, Parkville, Victoria 3010, Australia
| | - Anders Björklund
- Wallenberg Neuroscience Center, Department of Experimental Medical Science, Lund University, S-22184 Lund, Sweden.
| |
Collapse
|