1
|
Nagai H, Saito M, Iwata H. Direct conversion of urine-derived cells into functional motor neuron-like cells by defined transcription factors. Sci Rep 2024; 14:27011. [PMID: 39505927 PMCID: PMC11541886 DOI: 10.1038/s41598-024-73759-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Accepted: 09/20/2024] [Indexed: 11/08/2024] Open
Abstract
Direct cell-type conversion of somatic cells into cell types of interest has garnered great attention because it circumvents rejuvenation and preserves the hallmarks of cellular aging (unlike induced pluripotent stem cells [iPSCs]) and is more suitable for modeling diseases with strong age-related and epigenetic contributions. Fibroblasts are commonly used for direct conversion; however, obtaining these cells requires highly invasive skin biopsies. Urine-derived cells (UDCs) are an alternative cell source and can be obtained via noninvasive procedures. Herein, induced motor neuron-like cells (iMNs) were generated from UDCs by transducing transcription factors involved in motor neuron (MN) differentiation. iMNs exhibited neuronal morphology, upregulation of pan-neuron and MN markers, and MN functionality, including spontaneous calcium oscillation and bungarotoxin-positive neuromuscular junction formation, when co-cultured with myotubes. Altogether, the findings of this study indicated that UDCs can be converted to functional MNs. This technology may allow us to understand disease pathogenesis and progression and discover biomarkers and drugs for MN-related diseases at the population level.
Collapse
Affiliation(s)
- Hiroaki Nagai
- Neuroscience Drug Discovery Unit, Research, Takeda Pharmaceutical Company Limited, Fujisawa, 251-8555, Kanagawa, Japan.
| | - Masayo Saito
- Neuroscience Drug Discovery Unit, Research, Takeda Pharmaceutical Company Limited, Fujisawa, 251-8555, Kanagawa, Japan
| | - Hidehisa Iwata
- Neuroscience Drug Discovery Unit, Research, Takeda Pharmaceutical Company Limited, Fujisawa, 251-8555, Kanagawa, Japan.
| |
Collapse
|
2
|
Xu Z, Liu S, Xue X, Li W, Fu J, Deng CX. Rapid responses of human pluripotent stem cells to cyclic mechanical strains applied to integrin by acoustic tweezing cytometry. Sci Rep 2023; 13:18030. [PMID: 37865697 PMCID: PMC10590420 DOI: 10.1038/s41598-023-45397-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Accepted: 10/19/2023] [Indexed: 10/23/2023] Open
Abstract
Acoustic tweezing cytometry (ATC) is an ultrasound-based biophysical technique that has shown the capability to promote differentiation of human pluripotent stem cells (hPSCs). This study systematically examined how hPSCs respond to cyclic mechanical strains applied by ATC via displacement of integrin-bound microbubbles (averaged diameter of 4.3 µm) using ultrasound pulses (acoustic pressure 0.034 MPa, center frequency 1.24 MHz and pulse repetition frequency 1 Hz). Our data show downregulation of pluripotency marker Octamer-binding transcription factor 4 (OCT4) by at least 10% and increased nuclear localization of Yes-associated protein (YAP) by almost 100% in hPSCs immediately after ATC application for as short as 1 min and 5 min respectively. Analysis of the movements of integrin-anchored microbubbles under ATC stimulations reveals different stages of viscoelastic characteristic behavior and increasing deformation of the integrin-cytoskeleton (CSK) linkage. The peak displacement of integrin-bound microbubbles increased from 1.45 ± 0.16 to 4.74 ± 0.67 μm as the duty cycle of ultrasound pulses increased from 5% to 50% or the duration of each ultrasound pulse increased from 0.05 to 0.5 s. Real-time tracking of integrin-bound microbubbles during ATC application detects high correlation of microbubble displacements with OCT4 downregulation in hPSCs. Together, our data showing fast downregulation of OCT4 in hPSCs in respond to ATC stimulations highlight the unique mechanosensitivity of hPSCs to integrin-targeted cyclic force/strain dependent on the pulse duration or duty cycle of ultrasound pulses, providing insights into the mechanism of ATC-induced accelerated differentiation of hPSCs.
Collapse
Affiliation(s)
- Zhaoyi Xu
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Shiying Liu
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Xufeng Xue
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Weiping Li
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Jianping Fu
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA.
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA.
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI, 48109, USA.
| | - Cheri X Deng
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA.
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA.
| |
Collapse
|
3
|
Cheesbrough A, Harley P, Riccio F, Wu L, Song W, Lieberam I. A scalable human iPSC-based neuromuscular disease model on suspended biobased elastomer nanofiber scaffolds. Biofabrication 2023; 15:045020. [PMID: 37619554 PMCID: PMC10478173 DOI: 10.1088/1758-5090/acf39e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Revised: 08/05/2023] [Accepted: 08/24/2023] [Indexed: 08/26/2023]
Abstract
Many devastating neuromuscular diseases currently lack effective treatments. This is in part due to a lack of drug discovery platforms capable of assessing complex human neuromuscular disease phenotypes in a scalable manner. A major obstacle has been generating scaffolds to stabilise mature contractile myofibers in a multi-well assay format amenable to high content image (HCI) analysis. This study describes the development of a scalable human induced pluripotent stem cell (iPSC)-neuromuscular disease model, whereby suspended elastomer nanofibers support long-term stability, alignment, maturation, and repeated contractions of iPSC-myofibers, innervated by iPSC-motor neurons in 96-well assay plates. In this platform, optogenetic stimulation of the motor neurons elicits robust myofiber-contractions, providing a functional readout of neuromuscular transmission. Additionally, HCI analysis provides rapid and automated quantification of axonal outgrowth, myofiber morphology, and neuromuscular synapse number and morphology. By incorporating amyotrophic lateral sclerosis (ALS)-related TDP-43G298Smutant motor neurons and CRISPR-corrected controls, key neuromuscular disease phenotypes are recapitulated, including weaker myofiber contractions, reduced axonal outgrowth, and reduced number of neuromuscular synapses. Treatment with a candidate ALS drug, the receptor-interacting protein kinase-1 (RIPK1)-inhibitor necrostatin-1, rescues these phenotypes in a dose-dependent manner, highlighting the potential of this platform to screen novel treatments for neuromuscular diseases.
Collapse
Affiliation(s)
- Aimee Cheesbrough
- Centre for Gene Therapy & Regenerative Medicine, Faculty of Life Sciences & Medicine, King’s College London, London SE1 9RT, United Kingdom
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London SE1 1UL, United Kingdom
- UCL Centre for Biomaterials in Surgical Reconstruction and Regeneration, Department of Surgical Biotechnology, Division of Surgery and Interventional Science, University College London, London NW3 2PF, United Kingdom
| | - Peter Harley
- Centre for Gene Therapy & Regenerative Medicine, Faculty of Life Sciences & Medicine, King’s College London, London SE1 9RT, United Kingdom
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London SE1 1UL, United Kingdom
| | - Federica Riccio
- Centre for Gene Therapy & Regenerative Medicine, Faculty of Life Sciences & Medicine, King’s College London, London SE1 9RT, United Kingdom
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London SE1 1UL, United Kingdom
| | - Lei Wu
- UCL Centre for Biomaterials in Surgical Reconstruction and Regeneration, Department of Surgical Biotechnology, Division of Surgery and Interventional Science, University College London, London NW3 2PF, United Kingdom
| | - Wenhui Song
- UCL Centre for Biomaterials in Surgical Reconstruction and Regeneration, Department of Surgical Biotechnology, Division of Surgery and Interventional Science, University College London, London NW3 2PF, United Kingdom
| | - Ivo Lieberam
- Centre for Gene Therapy & Regenerative Medicine, Faculty of Life Sciences & Medicine, King’s College London, London SE1 9RT, United Kingdom
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London SE1 1UL, United Kingdom
| |
Collapse
|
4
|
Ashraf SS, Hosseinpour Sarmadi V, Larijani G, Naderi Garahgheshlagh S, Ramezani S, Moghadamifar S, Mohebi SL, Brouki Milan P, Haramshahi SMA, Ahmadirad N, Amini N. Regenerative medicine improve neurodegenerative diseases. Cell Tissue Bank 2023; 24:639-650. [PMID: 36527565 DOI: 10.1007/s10561-022-10062-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2022] [Accepted: 12/04/2022] [Indexed: 12/23/2022]
Abstract
Regenerative medicine is a subdivision of medicine that improves methods to regrow, repair or replace unhealthy cells and tissues to return to normal function. Cell therapy, gene therapy, nanomedicine as choices used to cure neurodegenerative disease. Recently, studies related to the treatment of neurodegenerative disorders have been focused on stem cell therapy and Nano-drugs beyond other than regenerative medicine. Hence, by data from experimental models and clinical trials, we review the impact of stem cell therapy, gene therapy, and nanomedicine on the treatment of Alzheimer's disease (AD), Parkinson's disease (PD), and Amyotrophic lateral sclerosis (ALS). Indeed, improved knowledge and continued research on gene therapy and nanomedicine in treating Alzheimer's disease, Parkinson's disease, and Amyotrophic lateral sclerosis lead to advancements in effective and practical treatments for neurodegenerative diseases.
Collapse
Affiliation(s)
- Seyedeh Sara Ashraf
- Department of Biology, Science and Research Branch, Islamic Azad University, Tehran, Iran
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran
- Institutes of Regenerative Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Vahid Hosseinpour Sarmadi
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran
- Institutes of Regenerative Medicine, Iran University of Medical Sciences, Tehran, Iran
- Department of Tissue Engineering and Regenerative Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Ghazaleh Larijani
- Department of Biology, Science and Research Branch, Islamic Azad University, Tehran, Iran
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Soheila Naderi Garahgheshlagh
- Burn Research Center, Iran University of Medical Sciences, Tehran, Iran
- Department of Plastic and Reconstructive surgery, Hazrat Fatemeh Hospital, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Sara Ramezani
- Neuroscience Research Center, Guilan Road Trauma Research Center, Guilan University of Medical Sciences, Rasht, Guilan, Iran
- Guilan Road Trauma Research Center, Guilan University of Medical Sciences, Rasht, Iran
| | - Soraya Moghadamifar
- Department of Textile Engineering, Amirkabir University of Technology, Tehran, Iran
| | - Seyedeh Lena Mohebi
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran
- Institutes of Regenerative Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Peiman Brouki Milan
- Institutes of Regenerative Medicine, Iran University of Medical Sciences, Tehran, Iran
- Department of Tissue Engineering and Regenerative Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Seyed Mohammad Amin Haramshahi
- Department of Tissue Engineering and Regenerative Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
- Department of Plastic and Reconstructive surgery, Hazrat Fatemeh Hospital, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Nooshin Ahmadirad
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran.
| | - Naser Amini
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran.
- Institutes of Regenerative Medicine, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
5
|
Patel T, Hammelman J, Aziz S, Jang S, Closser M, Michaels TL, Blum JA, Gifford DK, Wichterle H. Transcriptional dynamics of murine motor neuron maturation in vivo and in vitro. Nat Commun 2022; 13:5427. [PMID: 36109497 PMCID: PMC9477853 DOI: 10.1038/s41467-022-33022-4] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Accepted: 08/25/2022] [Indexed: 12/03/2022] Open
Abstract
Neurons born in the embryo can undergo a protracted period of maturation lasting well into postnatal life. How gene expression changes are regulated during maturation and whether they can be recapitulated in cultured neurons remains poorly understood. Here, we show that mouse motor neurons exhibit pervasive changes in gene expression and accessibility of associated regulatory regions from embryonic till juvenile age. While motifs of selector transcription factors, ISL1 and LHX3, are enriched in nascent regulatory regions, motifs of NFI factors, activity-dependent factors, and hormone receptors become more prominent in maturation-dependent enhancers. Notably, stem cell-derived motor neurons recapitulate ~40% of the maturation expression program in vitro, with neural activity playing only a modest role as a late-stage modulator. Thus, the genetic maturation program consists of a core hardwired subprogram that is correctly executed in vitro and an extrinsically-controlled subprogram that is dependent on the in vivo context of the maturing organism.
Collapse
Affiliation(s)
- Tulsi Patel
- Departments of Pathology & Cell Biology, Neuroscience, and Neurology, Columbia University Irving Medical Center, New York, NY, 10032, USA.
| | - Jennifer Hammelman
- Computer Science and Artificial Intelligence Laboratory, MIT, Cambridge, MA, 02139, USA
| | - Siaresh Aziz
- Departments of Pathology & Cell Biology, Neuroscience, and Neurology, Columbia University Irving Medical Center, New York, NY, 10032, USA
| | - Sumin Jang
- Departments of Pathology & Cell Biology, Neuroscience, and Neurology, Columbia University Irving Medical Center, New York, NY, 10032, USA
| | - Michael Closser
- Departments of Pathology & Cell Biology, Neuroscience, and Neurology, Columbia University Irving Medical Center, New York, NY, 10032, USA
| | - Theodore L Michaels
- Departments of Pathology & Cell Biology, Neuroscience, and Neurology, Columbia University Irving Medical Center, New York, NY, 10032, USA
| | - Jacob A Blum
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| | - David K Gifford
- Computer Science and Artificial Intelligence Laboratory, MIT, Cambridge, MA, 02139, USA
| | - Hynek Wichterle
- Departments of Pathology & Cell Biology, Neuroscience, and Neurology, Columbia University Irving Medical Center, New York, NY, 10032, USA.
| |
Collapse
|
6
|
Olmsted ZT, Stigliano C, Marzullo B, Cibelli J, Horner PJ, Paluh JL. Fully Characterized Mature Human iPS- and NMP-Derived Motor Neurons Thrive Without Neuroprotection in the Spinal Contusion Cavity. Front Cell Neurosci 2022; 15:725195. [PMID: 35046774 PMCID: PMC8762343 DOI: 10.3389/fncel.2021.725195] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Accepted: 11/04/2021] [Indexed: 11/21/2022] Open
Abstract
Neural cell interventions in spinal cord injury (SCI) have focused predominantly on transplanted multipotent neural stem/progenitor cells (NSPCs) for animal research and clinical use due to limited information on survival of spinal neurons. However, transplanted NSPC fate is unpredictable and largely governed by injury-derived matrix and cytokine factors that are often gliogenic and inflammatory. Here, using a rat cervical hemicontusion model, we evaluate the survival and integration of hiPSC-derived spinal motor neurons (SMNs) and oligodendrocyte progenitor cells (OPCs). SMNs and OPCs were differentiated in vitro through a neuromesodermal progenitor stage to mimic the natural origin of the spinal cord. We demonstrate robust survival and engraftment without additional injury site modifiers or neuroprotective biomaterials. Ex vivo differentiated neurons achieve cervical spinal cord matched transcriptomic and proteomic profiles, meeting functional electrophysiology parameters prior to transplantation. These data establish an approach for ex vivo developmentally accurate neuronal fate specification and subsequent transplantation for a more streamlined and predictable outcome in neural cell-based therapies of SCI.
Collapse
Affiliation(s)
- Zachary T. Olmsted
- Nanobioscience Constellation, Colleges of Nanoscale Science and Engineering, State University of New York Polytechnic Institute, Albany, NY, United States
| | - Cinzia Stigliano
- Center for Neuroregeneration, Department of Neurosurgery, Houston Methodist Research Institute, Houston, TX, United States
| | - Brandon Marzullo
- SUNY Buffalo Genomics and Bioinformatics Core, New York State Center of Excellence in Bioinformatics and Life Sciences, Buffalo, NY, United States
| | - Jose Cibelli
- Department of Animal Science, College of Agriculture and Natural Resources, Michigan State University, East Lansing, MI, United States
- Department of Large Animal Clinical Sciences, College of Veterinary Medicine, Michigan State University, East Lansing, MI, United States
| | - Philip J. Horner
- Center for Neuroregeneration, Department of Neurosurgery, Houston Methodist Research Institute, Houston, TX, United States
| | - Janet L. Paluh
- Nanobioscience Constellation, Colleges of Nanoscale Science and Engineering, State University of New York Polytechnic Institute, Albany, NY, United States
- *Correspondence: Janet L. Paluh
| |
Collapse
|
7
|
Abstract
Motoneurons are the 'final common path' between the central nervous system (that intends, selects, commands, and organises movement) and muscles (that produce the behaviour). Motoneurons are not passive relays, but rather integrate synaptic activity to appropriately tune output (spike trains) and therefore the production of muscle force. In this chapter, we focus on studies of mammalian motoneurons, describing their heterogeneity whilst providing a brief historical account of motoneuron recording techniques. Next, we describe adult motoneurons in terms of their passive, transition, and active (repetitive firing) properties. We then discuss modulation of these properties by somatic (C-boutons) and dendritic (persistent inward currents) mechanisms. Finally, we briefly describe select studies of human motor unit physiology and relate them to findings from animal preparations discussed earlier in the chapter. This interphyletic approach to the study of motoneuron physiology is crucial to progress understanding of how these diverse neurons translate intention into behaviour.
Collapse
|
8
|
Gupta S, Butler SJ. Getting in touch with your senses: Mechanisms specifying sensory interneurons in the dorsal spinal cord. WIREs Mech Dis 2021; 13:e1520. [PMID: 34730293 PMCID: PMC8459260 DOI: 10.1002/wsbm.1520] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Revised: 01/10/2021] [Accepted: 01/11/2021] [Indexed: 11/18/2022]
Abstract
The spinal cord is functionally and anatomically divided into ventrally derived motor circuits and dorsally derived somatosensory circuits. Sensory stimuli originating either at the periphery of the body, or internally, are relayed to the dorsal spinal cord where they are processed by distinct classes of sensory dorsal interneurons (dIs). dIs convey sensory information, such as pain, heat or itch, either to the brain, and/or to the motor circuits to initiate the appropriate response. They also regulate the intensity of sensory information and are the major target for the opioid analgesics. While the developmental mechanisms directing ventral and dorsal cell fates have been hypothesized to be similar, more recent research has suggested that dI fates are specified by novel mechanisms. In this review, we will discuss the molecular events that specify dorsal neuronal patterning in the spinal cord, thereby generating diverse dI identities. We will then discuss how this molecular understanding has led to the development of robust stem cell methods to derive multiple spinal cell types, including the dIs, and the implication of these studies for treating spinal cord injuries and neurodegenerative diseases. This article is categorized under: Neurological Diseases > Stem Cells and Development.
Collapse
Affiliation(s)
- Sandeep Gupta
- Department of NeurobiologyUniversity of California, Los AngelesLos AngelesCaliforniaUSA
| | - Samantha J. Butler
- Department of NeurobiologyUniversity of California, Los AngelesLos AngelesCaliforniaUSA
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell ResearchUniversity of California, Los AngelesLos AngelesCaliforniaUSA
- Intellectual and Developmental Disabilities Research CenterUniversity of California, Los AngelesLos AngelesCaliforniaUSA
| |
Collapse
|
9
|
Iwata R. Temporal differences of neurodevelopment processes between species. Neurosci Res 2021; 177:8-15. [PMID: 34419562 DOI: 10.1016/j.neures.2021.08.004] [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: 03/30/2021] [Revised: 07/08/2021] [Accepted: 08/17/2021] [Indexed: 02/06/2023]
Abstract
The ontogeny programs are highly conserved across all vertebrates, although there are significant temporal variations in interspecies developmental processes. Changing the timing and rate of developmental processes could affect subsequent organogenesis profoundly and may also have been critical factors in evolutionary diversity. However, despite their potential importance, the cellular and molecular mechanisms that control interspecies differences in developmental timescale remain unclear. This review highlights recent advances in the experimental models to compare interspecies differences in neurodevelopmental processes, neurogenesis, and neuronal maturation and discusses the possible mechanisms that could generate species-specific timescales.
Collapse
Affiliation(s)
- Ryohei Iwata
- VIB KU Leuven Center for Brain & Disease Research, KU Leuven Department of Neurosciences, Leuven Brain Institute, 3000 Leuven, Belgium; Université Libre de Bruxelles (ULB), Institut de Recherches en Biologie Humaine et Moléculaire (IRIBHM), ULB Neuroscience Institute (UNI), 1070 Brussels, Belgium.
| |
Collapse
|
10
|
Nedelec S, Martinez-Arias A. In vitro models of spinal motor circuit's development in mammals: achievements and challenges. Curr Opin Neurobiol 2021; 66:240-249. [PMID: 33677159 DOI: 10.1016/j.conb.2020.12.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Revised: 10/12/2020] [Accepted: 12/02/2020] [Indexed: 12/11/2022]
Abstract
The connectivity patterns of neurons sustaining the functionality of spinal locomotor circuits rely on the specification of hundreds of motor neuron and interneuron subtypes precisely arrayed within the embryonic spinal cord. Knowledge acquired by developmental biologists on the molecular mechanisms underpinning this process in vivo has supported the development of 2D and 3D differentiation strategies to generate spinal neuronal diversity from mouse and human pluripotent stem cells (PSCs). Here, we review recent breakthroughs in this field and the perspectives opened up by models of in vitro embryogenesis to approach the mechanisms underlying neuronal diversification and the formation of functional mouse and human locomotor circuits. Beyond serving fundamental investigations, these new approaches should help engineering neuronal circuits differentially impacted in neuromuscular disorders, such as amyotrophic lateral sclerosis or spinal muscular atrophies, and thus open new avenues for disease modeling and drug screenings.
Collapse
Affiliation(s)
- Stéphane Nedelec
- Institut du Fer à Moulin, 75005, Paris, France; Inserm, UMR-S 1270, 75005 Paris, France; Sorbonne Université, Science and Engineering Faculty, 75005 Paris, France.
| | | |
Collapse
|
11
|
Smith CC, Brownstone RM. Spinal motoneuron firing properties mature from rostral to caudal during postnatal development of the mouse. J Physiol 2020; 598:5467-5485. [PMID: 32851667 PMCID: PMC8436765 DOI: 10.1113/jp280274] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Accepted: 08/24/2020] [Indexed: 12/16/2022] Open
Abstract
Key points Many mammals are born with immature motor systems that develop through a critical period of postnatal development. In rodents, postnatal maturation of movement occurs from rostral to caudal, correlating with maturation of descending supraspinal and local spinal circuits. We asked whether development of fundamental electrophysiological properties of spinal motoneurons follows the same rostro‐caudal sequence. We show that in both regions, repetitive firing parameters increase and excitability decreases with development; however, these characteristics mature earlier in cervical motoneurons. We suggest that in addition to autonomous mechanisms, motoneuron development depends on activity resulting from their circuit milieu.
Abstract Altricial mammals are born with immature nervous systems comprised of circuits that do not yet have the neuronal properties and connectivity required to produce future behaviours. During the critical period of postnatal development, neuronal properties are tuned to participate in functional circuits. In rodents, cervical motoneurons are born prior to lumbar motoneurons, and spinal cord development follows a sequential rostro‐caudal pattern. Here we asked whether birth order is reflected in the postnatal development of electrophysiological properties. We show that motoneurons of both regions have similar properties at birth and follow the same developmental profile, with maximal firing increasing and excitability decreasing into the third postnatal week. However, these maturative processes occur in cervical motoneurons prior to lumbar motoneurons, correlating with the maturation of premotor descending and local spinal systems. These results suggest that motoneuron properties do not mature by cell autonomous mechanisms alone, but also depend on developing premotor circuits. Many mammals are born with immature motor systems that develop through a critical period of postnatal development. In rodents, postnatal maturation of movement occurs from rostral to caudal, correlating with maturation of descending supraspinal and local spinal circuits. We asked whether development of fundamental electrophysiological properties of spinal motoneurons follows the same rostro‐caudal sequence. We show that in both regions, repetitive firing parameters increase and excitability decreases with development; however, these characteristics mature earlier in cervical motoneurons. We suggest that in addition to autonomous mechanisms, motoneuron development depends on activity resulting from their circuit milieu.
Collapse
Affiliation(s)
- Calvin C Smith
- Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, University College London, London, UK
| | - Robert M Brownstone
- Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, University College London, London, UK
| |
Collapse
|
12
|
Zhao AD, Qin H, Sun ML, Ma K, Fu XB. Efficient and rapid conversion of human astrocytes and ALS mouse model spinal cord astrocytes into motor neuron-like cells by defined small molecules. Mil Med Res 2020; 7:42. [PMID: 32892745 PMCID: PMC7487818 DOI: 10.1186/s40779-020-00271-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Accepted: 08/27/2020] [Indexed: 01/25/2023] Open
Abstract
BACKGROUND Motor neuron degeneration or loss in the spinal cord is the characteristic phenotype of motor neuron diseases or spinal cord injuries. Being proliferative and located near neurons, astrocytes are considered ideal cell sources for regenerating neurons. METHODS We selected and tested different combinations of the small molecules for inducing the conversion of human and mouse astrocytes into neurons. Microscopic imaging and immunocytochemistry analyses were used to characterize the morphology and phenotype of the induced neurons while RT-qPCR was utilized to analyze changes in gene expression. In addition, whole-cell patch-clamp recordings were measured to examine the electrophysiological properties of induced neurons. RESULTS The results showed that human astrocytes could be rapidly and efficiently converted into motor neuron-like cells by treatment with defined small molecules, with a yield of over 85% motor neuron-like cells attained. The induced motor neuron-like cells expressed the pan-neuronal markers TUJ1, MAP2, NeuN, and Synapsin 1 and motor neuron markers HB9, ISL1, CHAT, and VAChT. During the conversion process, the cells did not pass through a proliferative neural progenitor cell intermediate. The induced motor neurons were functional, showing the electrophysiological properties of neurons. The same chemical cocktail could induce spinal cord astrocytes from an amyotrophic lateral sclerosis mouse model carrying a SOD1 mutation to become motor neuron-like cells that exhibited a decrease in cell survival and an increase in oxidative stress compared to that observed in wild-type MNs derived from healthy mice. Moreover, the chemical induction reduced oxidative stress in the mutant astrocytes. CONCLUSION The results of the present study demonstrated the feasibility of chemically converting human and mouse astrocytes into motor neuron-like cells that are useful for neurodegenerative disease modeling and regenerative medicine.
Collapse
Affiliation(s)
- An-Dong Zhao
- Tianjin Medical University, Tianjin, 300070, China.,Research Center for Tissue Repair and Regeneration affiliated to the Medical Innovation Research Division and 4th Medical Center, PLA General Hospital and PLA Medical College, 28 Fu Xing Road, Haidian District, Beijing, 100853, P. R. China
| | - Hua Qin
- Research Center for Tissue Repair and Regeneration affiliated to the Medical Innovation Research Division and 4th Medical Center, PLA General Hospital and PLA Medical College, 28 Fu Xing Road, Haidian District, Beijing, 100853, P. R. China.,PLA Key Laboratory of Tissue Repair and Regenerative Medicine and Beijing Key Research Laboratory of Skin Injury, Repair and Regeneration, Beijing, 100048, China
| | - Meng-Li Sun
- Research Center for Tissue Repair and Regeneration affiliated to the Medical Innovation Research Division and 4th Medical Center, PLA General Hospital and PLA Medical College, 28 Fu Xing Road, Haidian District, Beijing, 100853, P. R. China.,PLA Key Laboratory of Tissue Repair and Regenerative Medicine and Beijing Key Research Laboratory of Skin Injury, Repair and Regeneration, Beijing, 100048, China
| | - Kui Ma
- Research Center for Tissue Repair and Regeneration affiliated to the Medical Innovation Research Division and 4th Medical Center, PLA General Hospital and PLA Medical College, 28 Fu Xing Road, Haidian District, Beijing, 100853, P. R. China.,PLA Key Laboratory of Tissue Repair and Regenerative Medicine and Beijing Key Research Laboratory of Skin Injury, Repair and Regeneration, Beijing, 100048, China
| | - Xiao-Bing Fu
- Research Center for Tissue Repair and Regeneration affiliated to the Medical Innovation Research Division and 4th Medical Center, PLA General Hospital and PLA Medical College, 28 Fu Xing Road, Haidian District, Beijing, 100853, P. R. China. .,PLA Key Laboratory of Tissue Repair and Regenerative Medicine and Beijing Key Research Laboratory of Skin Injury, Repair and Regeneration, Beijing, 100048, China. .,Research Unit of Trauma Care, Tissue Repair and Regeneration, Chinese Academy of Medical Sciences, Beijing, 100048, China.
| |
Collapse
|
13
|
Optimized Protocol to Generate Spinal Motor Neuron Cells from Induced Pluripotent Stem Cells from Charcot Marie Tooth Patients. Brain Sci 2020; 10:brainsci10070407. [PMID: 32605002 PMCID: PMC7408498 DOI: 10.3390/brainsci10070407] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Revised: 06/18/2020] [Accepted: 06/24/2020] [Indexed: 01/09/2023] Open
Abstract
Modelling rare neurogenetic diseases to develop new therapeutic strategies is highly challenging. The use of human-induced pluripotent stem cells (hiPSCs) is a powerful approach to obtain specialized cells from patients. For hereditary peripheral neuropathies, such as Charcot–Marie–Tooth disease (CMT) Type II, spinal motor neurons (MNs) are impaired but are very difficult to study. Although several protocols are available to differentiate hiPSCs into neurons, their efficiency is still poor for CMT patients. Thus, our goal was to develop a robust, easy, and reproducible protocol to obtain MNs from CMT patient hiPSCs. The presented protocol generates MNs within 20 days, with a success rate of 80%, using specifically chosen molecules, such as Sonic Hedgehog or retinoic acid. The timing and concentrations of the factors used to induce differentiation are crucial and are given hereby. We then assessed the MNs by optic microscopy, immunocytochemistry (Islet1/2, HB9, Tuj1, and PGP9.5), and electrophysiological recordings. This method of generating MNs from CMT patients in vitro shows promise for the further development of assays to understand the pathological mechanisms of CMT and for drug screening.
Collapse
|
14
|
Bubnys A, Kandel H, Kao LM, Pfaff D, Tabansky I. Hindbrain V2a Neurons Pattern Rhythmic Activity of Motor Neurons in a Reticulospinal Coculture. Front Neurosci 2019; 13:1077. [PMID: 31680817 PMCID: PMC6811747 DOI: 10.3389/fnins.2019.01077] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Accepted: 09/24/2019] [Indexed: 11/19/2022] Open
Abstract
As the capacity to isolate distinct neuronal cell types has advanced over the past several decades, new two- and three-dimensional in vitro models of the interactions between different brain regions have expanded our understanding of human neurobiology and the origins of disease. These cultures develop distinctive patterns of activity, but the extent that these patterns are determined by the molecular identity of individual cell types versus the specific pattern of network connectivity is unclear. To address the question of how individual cell types interact in vitro, we developed a simplified culture using two excitatory neuronal subtypes known to participate in the in vivo reticulospinal circuit: HB9+ spinal motor neurons and Chx10+ hindbrain V2a neurons. Here, we report the emergence of cell type-specific patterns of activity in culture; on their own, Chx10+ neurons developed regular, synchronized bursts of activity that recruited neurons across the entire culture, whereas HB9+ neuron activity consisted of an irregular pattern. When these two subtypes were cocultured, HB9+ neurons developed synchronized network bursts that were precisely correlated with Chx10+ neuron activity, thereby recreating an aspect of Chx10+ neurons' role in driving motor activity. These bursts were dependent on AMPA receptors. Our results demonstrate that the molecular classification of the neurons comprising in vitro networks is a crucial determinant of their activity. It is therefore possible to improve both the reproducibility and the applicability of in vitro neurobiological and disease models by carefully controlling the constituent mixtures of neuronal subtypes.
Collapse
Affiliation(s)
- Adele Bubnys
- Laboratory of Neurobiology and Behavior, The Rockefeller University, New York, NY, United States
| | - Hagar Kandel
- Laboratory of Neurobiology and Behavior, The Rockefeller University, New York, NY, United States
| | - Lee Ming Kao
- Laboratory of Neurobiology and Behavior, The Rockefeller University, New York, NY, United States
| | - Donald Pfaff
- Laboratory of Neurobiology and Behavior, The Rockefeller University, New York, NY, United States
| | - Inna Tabansky
- Laboratory of Neurobiology and Behavior, The Rockefeller University, New York, NY, United States
- Feinstein Institute for Medical Research, Manhasset, NY, United States
| |
Collapse
|
15
|
Aydin O, Zhang X, Nuethong S, Pagan-Diaz GJ, Bashir R, Gazzola M, Saif MTA. Neuromuscular actuation of biohybrid motile bots. Proc Natl Acad Sci U S A 2019; 116:19841-19847. [PMID: 31527266 PMCID: PMC6778261 DOI: 10.1073/pnas.1907051116] [Citation(s) in RCA: 75] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
The integration of muscle cells with soft robotics in recent years has led to the development of biohybrid machines capable of untethered locomotion. A major frontier that currently remains unexplored is neuronal actuation and control of such muscle-powered biohybrid machines. As a step toward this goal, we present here a biohybrid swimmer driven by on-board neuromuscular units. The body of the swimmer consists of a free-standing soft scaffold, skeletal muscle tissue, and optogenetic stem cell-derived neural cluster containing motor neurons. Myoblasts embedded in extracellular matrix self-organize into a muscle tissue guided by the geometry of the scaffold, and the resulting muscle tissue is cocultured in situ with a neural cluster. Motor neurons then extend neurites selectively toward the muscle and innervate it, developing functional neuromuscular units. Based on this initial construct, we computationally designed, optimized, and implemented light-sensitive flagellar swimmers actuated by these neuromuscular units. Cyclic muscle contractions, induced by neural stimulation, drive time-irreversible flagellar dynamics, thereby providing thrust for untethered forward locomotion of the swimmer. Overall, this work demonstrates an example of a biohybrid robot implementing neuromuscular actuation and illustrates a path toward the forward design and control of neuron-enabled biohybrid machines.
Collapse
Affiliation(s)
- Onur Aydin
- Department of Mechanical Science and Engineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801
| | - Xiaotian Zhang
- Department of Mechanical Science and Engineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801
| | - Sittinon Nuethong
- Department of Mechanical Science and Engineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801
| | - Gelson J Pagan-Diaz
- Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801
| | - Rashid Bashir
- Department of Mechanical Science and Engineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801
- Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801
| | - Mattia Gazzola
- Department of Mechanical Science and Engineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801;
- National Center for Supercomputing Applications, University of Illinois at Urbana-Champaign, Urbana, IL 61801
| | - M Taher A Saif
- Department of Mechanical Science and Engineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801;
- Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801
| |
Collapse
|
16
|
Machado CB, Pluchon P, Harley P, Rigby M, Gonzalez Sabater V, Stevenson DC, Hynes S, Lowe A, Burrone J, Viasnoff V, Lieberam I. In Vitro Modelling of Nerve-Muscle Connectivity in a Compartmentalised Tissue Culture Device. ADVANCED BIOSYSTEMS 2019; 3:1800307. [PMID: 31428672 PMCID: PMC6699992 DOI: 10.1002/adbi.201800307] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/17/2018] [Indexed: 01/02/2023]
Abstract
Motor neurons project axons from the hindbrain and spinal cord to muscle, where they induce myofibre contractions through neurotransmitter release at neuromuscular junctions. Studies of neuromuscular junction formation and homeostasis have been largely confined to in vivo models. In this study we have merged three powerful tools - pluripotent stem cells, optogenetics and microfabrication - and designed an open microdevice in which motor axons grow from a neural compartment containing embryonic stem cell-derived motor neurons and astrocytes through microchannels to form functional neuromuscular junctions with contractile myofibers in a separate compartment. Optogenetic entrainment of motor neurons in this reductionist neuromuscular circuit enhanced neuromuscular junction formation more than two-fold, mirroring the activity-dependence of synapse development in vivo. We incorporated an established motor neuron disease model into our system and found that coculture of motor neurons with SOD1G93A astrocytes resulted in denervation of the central compartment and diminished myofiber contractions, a phenotype which was rescued by the Receptor Interacting Serine/Threonine Kinase 1 (RIPK1) inhibitor Necrostatin. This coculture system replicates key aspects of nerve-muscle connectivity in vivo and represents a rapid and scalable alternative to animal models of neuromuscular function and disease.
Collapse
Affiliation(s)
- Carolina Barcellos Machado
- Centre for Stem Cells and Regenerative Medicine, King’s
College London, London SE1 9RT, UK; Centre for Developmental
Neurobiology/MRC Centre for Neurodevelopmental Disorders, King’s
College London, London SE1 1UL, UK
| | - Perrine Pluchon
- Centre for Stem Cells and Regenerative Medicine, King’s
College London, London SE1 9RT, UK; Centre for Developmental
Neurobiology/MRC Centre for Neurodevelopmental Disorders, King’s
College London, London SE1 1UL, UK; Mechanobiology Institute, National
University of Singapore, Singapore 117411
| | - Peter Harley
- Centre for Stem Cells and Regenerative Medicine, King’s College London, London SE1 9RT, UK; Centre for Developmental Neurobiology/MRC Centre for Neurodevelopmental Disorders, King’s College London, London SE1 1UL, UK
| | | | - Victoria Gonzalez Sabater
- Centre for Developmental Neurobiology/MRC Centre for Neurodevelopmental Disorders, King’s College London, London SE1 1UL, UK
| | | | - Stephanie Hynes
- Centre for Stem Cells and Regenerative Medicine, King’s College London, London SE1 9RT, UK; Centre for Developmental Neurobiology/MRC Centre for Neurodevelopmental Disorders, King’s College London, London SE1 1UL, UK
| | - Andrew Lowe
- Centre for Developmental Neurobiology, King’s College London, London SE1 1UL, UK
| | - Juan Burrone
- Centre for Developmental Neurobiology/MRC Centre for Neurodevelopmental Disorders, King’s College London, London SE1 1UL, UK
| | - Virgile Viasnoff
- Mechanobiology Institute, National University of Singapore,
Singapore 117411
| | - Ivo Lieberam
- Centre for Stem Cells and Regenerative Medicine, King’s
College London, London SE1 9RT, UK; Centre for Developmental
Neurobiology/MRC Centre for Neurodevelopmental Disorders, King’s
College London, London SE1 1UL, UK
| |
Collapse
|
17
|
Luo B, Tian L, Chen N, Ramakrishna S, Thakor N, Yang IH. Electrospun nanofibers facilitate better alignment, differentiation, and long-term culture in an in vitro model of the neuromuscular junction (NMJ). Biomater Sci 2019; 6:3262-3272. [PMID: 30402630 DOI: 10.1039/c8bm00720a] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
The neuromuscular junction (NMJ) is a specialized synapse between motor neurons and the muscle fibers they innervate. Due to the complexity of various signalling molecules and pathways, in vivo NMJs are difficult to study. Therefore, in vitro motor neuron-muscle co-culture plays a pivotal role in studying the mechanisms of NMJ formation associated with neurodegenerative diseases. There is a growing need to develop novel methodologies that can be used to develop long-term cultures of NMJs. To date, there have been few studies on NMJ development and long-term maintenance of the system, which is also the main challenge for the current in vitro models of NMJs. In this study, we demonstrate a long-term co-culture system of primary embryonic motor neurons from Sprague-Dawley rats and C2C12 cells on both random and aligned electrospun polylactic acid (PLA) nanofibrous scaffolds. This is the first study to explore the role of electrospun nanofibers in the long-term maintenance of NMJs. PLA nanofibrous scaffolds provide better contact guidance for C2C12 cells aligning along the fibers, thus guiding myotube formation. We can only maintain the co-culture system on a conventional glass substrate for 2 weeks, whilst 55% and 70% of the cells still survived on random and aligned PLA substrates after 7 weeks. Our nanofiber-based long-term co-culture system is used as an important tool for the fundamental research of NMJs.
Collapse
Affiliation(s)
- Baiwen Luo
- Singapore Institute for Neurotechnology, National University of Singapore, 28 Medical Drive, #05-COR, Singapore 119077. inhong.yang.@ku.ac.ae
| | | | | | | | | | | |
Collapse
|
18
|
Thams S, Lowry ER, Larraufie MH, Spiller KJ, Li H, Williams DJ, Hoang P, Jiang E, Williams LA, Sandoe J, Eggan K, Lieberam I, Kanning KC, Stockwell BR, Henderson CE, Wichterle H. A Stem Cell-Based Screening Platform Identifies Compounds that Desensitize Motor Neurons to Endoplasmic Reticulum Stress. Mol Ther 2019; 27:87-101. [PMID: 30446391 PMCID: PMC6318783 DOI: 10.1016/j.ymthe.2018.10.010] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2018] [Revised: 10/07/2018] [Accepted: 10/16/2018] [Indexed: 02/06/2023] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a neurodegenerative disease selectively targeting motor neurons in the brain and spinal cord. The reasons for differential motor neuron susceptibility remain elusive. We developed a stem cell-based motor neuron assay to study cell-autonomous mechanisms causing motor neuron degeneration, with implications for ALS. A small-molecule screen identified cyclopiazonic acid (CPA) as a stressor to which stem cell-derived motor neurons were more sensitive than interneurons. CPA induced endoplasmic reticulum stress and the unfolded protein response. Furthermore, CPA resulted in an accelerated degeneration of motor neurons expressing human superoxide dismutase 1 (hSOD1) carrying the ALS-causing G93A mutation, compared to motor neurons expressing wild-type hSOD1. A secondary screen identified compounds that alleviated CPA-mediated motor neuron degeneration: three kinase inhibitors and tauroursodeoxycholic acid (TUDCA), a bile acid derivative. The neuroprotective effects of these compounds were validated in human stem cell-derived motor neurons carrying a mutated SOD1 allele (hSOD1A4V). Moreover, we found that the administration of TUDCA in an hSOD1G93A mouse model of ALS reduced muscle denervation. Jointly, these results provide insights into the mechanisms contributing to the preferential susceptibility of ALS motor neurons, and they demonstrate the utility of stem cell-derived motor neurons for the discovery of new neuroprotective compounds.
Collapse
Affiliation(s)
- Sebastian Thams
- Department of Pathology and Cell Biology, Center for Motor Neuron Biology and Disease, Columbia Stem Cell Initiative, Columbia University Irving Medical Center, 630 West 168th Street, New York, NY 10032, USA.
| | - Emily Rhodes Lowry
- Department of Pathology and Cell Biology, Center for Motor Neuron Biology and Disease, Columbia Stem Cell Initiative, Columbia University Irving Medical Center, 630 West 168th Street, New York, NY 10032, USA
| | - Marie-Hélène Larraufie
- Department of Biological Sciences and Department of Chemistry, Columbia University, Northwest Corner Building, MC4846, 550 West 120th Street, New York, NY 10027, USA
| | - Krista J Spiller
- Department of Pathology and Cell Biology, Center for Motor Neuron Biology and Disease, Columbia Stem Cell Initiative, Columbia University Irving Medical Center, 630 West 168th Street, New York, NY 10032, USA
| | - Hai Li
- Department of Pathology and Cell Biology, Center for Motor Neuron Biology and Disease, Columbia Stem Cell Initiative, Columbia University Irving Medical Center, 630 West 168th Street, New York, NY 10032, USA
| | - Damian J Williams
- Department of Physiology and Cellular Biophysics, Columbia University Irving Medical Center, 650 West 168th Street, New York, NY, USA
| | - Phuong Hoang
- Department of Pathology and Cell Biology, Center for Motor Neuron Biology and Disease, Columbia Stem Cell Initiative, Columbia University Irving Medical Center, 630 West 168th Street, New York, NY 10032, USA
| | - Elise Jiang
- Department of Biological Sciences and Department of Chemistry, Columbia University, Northwest Corner Building, MC4846, 550 West 120th Street, New York, NY 10027, USA
| | - Luis A Williams
- Department of Stem Cell and Regenerative Biology, Harvard University, MA 02138, USA
| | - Jackson Sandoe
- Department of Stem Cell and Regenerative Biology, Harvard University, MA 02138, USA
| | - Kevin Eggan
- Department of Stem Cell and Regenerative Biology, Harvard University, MA 02138, USA
| | - Ivo Lieberam
- Centre for Stem Cells and Regenerative Medicine and MRC Centre for Neurodevelopmental Disorders, King's College London, London SE1 9RT, UK
| | - Kevin C Kanning
- Department of Pathology and Cell Biology, Center for Motor Neuron Biology and Disease, Columbia Stem Cell Initiative, Columbia University Irving Medical Center, 630 West 168th Street, New York, NY 10032, USA
| | - Brent R Stockwell
- Department of Biological Sciences and Department of Chemistry, Columbia University, Northwest Corner Building, MC4846, 550 West 120th Street, New York, NY 10027, USA
| | - Christopher E Henderson
- Department of Pathology and Cell Biology, Center for Motor Neuron Biology and Disease, Columbia Stem Cell Initiative, Columbia University Irving Medical Center, 630 West 168th Street, New York, NY 10032, USA
| | - Hynek Wichterle
- Department of Pathology and Cell Biology, Center for Motor Neuron Biology and Disease, Columbia Stem Cell Initiative, Columbia University Irving Medical Center, 630 West 168th Street, New York, NY 10032, USA; Departments of Neuroscience, Rehabilitation and Regenerative Medicine, and Neurology, Columbia University Irving Medical Center, 630 West 168th Street, New York, NY 10032, USA.
| |
Collapse
|
19
|
Abd Al Samid M, McPhee JS, Saini J, McKay TR, Fitzpatrick LM, Mamchaoui K, Bigot A, Mouly V, Butler-Browne G, Al-Shanti N. A functional human motor unit platform engineered from human embryonic stem cells and immortalized skeletal myoblasts. STEM CELLS AND CLONING-ADVANCES AND APPLICATIONS 2018; 11:85-93. [PMID: 30519053 PMCID: PMC6233953 DOI: 10.2147/sccaa.s178562] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Background Although considerable research on neuromuscular junctions (NMJs) has been conducted, the prospect of in vivo NMJ studies is limited and these studies are challenging to implement. Therefore, there is a clear unmet need to develop a feasible, robust, and physiologically relevant in vitro NMJ model. Objective We aimed to establish a novel functional human NMJs platform, which is serum and neural complex media/neural growth factor-free, using human immortalized myoblasts and human embryonic stem cells (hESCs)-derived neural progenitor cells (NPCs) that can be used to understand the mechanisms of NMJ development and degeneration. Methods Immortalized human myoblasts were co-cultured with hESCs derived committed NPCs. Over the course of the 7 days myoblasts differentiated into myotubes and NPCs differentiated into motor neurons. Results Neuronal axon sprouting branched to form multiple NMJ innervation sites along the myotubes and the myotubes showed extensive, spontaneous contractile activity. Choline acetyltransferase and βIII-tubulin immunostaining confirmed that the NPCs had matured into cholinergic motor neurons. Postsynaptic site of NMJs was further characterized by staining dihydropyridine receptors, ryanodine receptors, and acetylcholine receptors by α-bungarotoxin. Conclusion We established a functional human motor unit platform for in vitro investigations. Thus, this co-culture system can be used as a novel platform for 1) drug discovery in the treatment of neuromuscular disorders, 2) deciphering vital features of NMJ formation, regulation, maintenance, and repair, and 3) exploring neuromuscular diseases, age-associated degeneration of the NMJ, muscle aging, and diabetic neuropathy and myopathy.
Collapse
Affiliation(s)
- Marwah Abd Al Samid
- Healthcare Science Research Institute, School of Healthcare Science, Manchester Metropolitan University, Manchester, UK,
| | - Jamie S McPhee
- Department of Sport and Exercise Science, Manchester Metropolitan University, Manchester, UK
| | - Jasdeep Saini
- Healthcare Science Research Institute, School of Healthcare Science, Manchester Metropolitan University, Manchester, UK,
| | - Tristan R McKay
- Healthcare Science Research Institute, School of Healthcare Science, Manchester Metropolitan University, Manchester, UK,
| | - Lorna M Fitzpatrick
- Healthcare Science Research Institute, School of Healthcare Science, Manchester Metropolitan University, Manchester, UK,
| | - Kamel Mamchaoui
- Center for Research in Myology, Sorbonne Université-INSERM, Paris, France
| | - Anne Bigot
- Center for Research in Myology, Sorbonne Université-INSERM, Paris, France
| | - Vincent Mouly
- Center for Research in Myology, Sorbonne Université-INSERM, Paris, France
| | | | - Nasser Al-Shanti
- Healthcare Science Research Institute, School of Healthcare Science, Manchester Metropolitan University, Manchester, UK,
| |
Collapse
|
20
|
De Santis R, Garone MG, Pagani F, de Turris V, Di Angelantonio S, Rosa A. Direct conversion of human pluripotent stem cells into cranial motor neurons using a piggyBac vector. Stem Cell Res 2018; 29:189-196. [DOI: 10.1016/j.scr.2018.04.012] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/31/2017] [Revised: 03/23/2018] [Accepted: 04/24/2018] [Indexed: 12/11/2022] Open
|
21
|
Jacko M, Weyn-Vanhentenryck SM, Smerdon JW, Yan R, Feng H, Williams DJ, Pai J, Xu K, Wichterle H, Zhang C. Rbfox Splicing Factors Promote Neuronal Maturation and Axon Initial Segment Assembly. Neuron 2018; 97:853-868.e6. [PMID: 29398366 PMCID: PMC5823762 DOI: 10.1016/j.neuron.2018.01.020] [Citation(s) in RCA: 88] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2017] [Revised: 08/28/2017] [Accepted: 01/08/2018] [Indexed: 12/22/2022]
Abstract
Neuronal maturation requires dramatic morphological and functional changes, but the molecular mechanisms governing this process are not well understood. Here, we studied the role of Rbfox1, Rbfox2, and Rbfox3 proteins, a family of tissue-specific splicing regulators mutated in multiple neurodevelopmental disorders. We generated Rbfox triple knockout (tKO) ventral spinal neurons to define a comprehensive network of alternative exons under Rbfox regulation and to investigate their functional importance in the developing neurons. Rbfox tKO neurons exhibit defects in alternative splicing of many cytoskeletal, membrane, and synaptic proteins, and display immature electrophysiological activity. The axon initial segment (AIS), a subcellular structure important for action potential initiation, is diminished upon Rbfox depletion. We identified an Rbfox-regulated splicing switch in ankyrin G, the AIS "interaction hub" protein, that regulates ankyrin G-beta spectrin affinity and AIS assembly. Our data show that the Rbfox-regulated splicing program plays a crucial role in structural and functional maturation of postmitotic neurons.
Collapse
Affiliation(s)
- Martin Jacko
- Departments of Systems Biology and Biochemistry and Molecular Biophysics, Center for Motor Neuron Biology and Disease, Columbia University, New York, NY 10032, USA; Departments of Pathology and Cell Biology, Neurology, and Neuroscience, Center for Motor Neuron Biology and Disease, Columbia Stem Cell Initiative, Columbia University, New York, NY 10032, USA
| | - Sebastien M Weyn-Vanhentenryck
- Departments of Systems Biology and Biochemistry and Molecular Biophysics, Center for Motor Neuron Biology and Disease, Columbia University, New York, NY 10032, USA
| | - John W Smerdon
- Departments of Pathology and Cell Biology, Neurology, and Neuroscience, Center for Motor Neuron Biology and Disease, Columbia Stem Cell Initiative, Columbia University, New York, NY 10032, USA; Department of Physiology and Cellular Biophysics, Columbia University, New York, NY 10032, USA
| | - Rui Yan
- Department of Chemistry, University of California, Berkeley, Berkeley, CA 94720, USA; Chan Zuckerberg Biohub, San Francisco, CA 94158, USA
| | - Huijuan Feng
- Departments of Systems Biology and Biochemistry and Molecular Biophysics, Center for Motor Neuron Biology and Disease, Columbia University, New York, NY 10032, USA; MOE Key Laboratory of Bioinformatics and Bioinformatics Division, TNLIST/Department of Automation, Tsinghua University, Beijing 100084, China
| | - Damian J Williams
- Columbia University Stem Cell Core Facility, Department of Rehabilitation and Regenerative Medicine, New York, NY 10032, USA
| | - Joy Pai
- Departments of Systems Biology and Biochemistry and Molecular Biophysics, Center for Motor Neuron Biology and Disease, Columbia University, New York, NY 10032, USA
| | - Ke Xu
- Department of Chemistry, University of California, Berkeley, Berkeley, CA 94720, USA; Chan Zuckerberg Biohub, San Francisco, CA 94158, USA
| | - Hynek Wichterle
- Departments of Pathology and Cell Biology, Neurology, and Neuroscience, Center for Motor Neuron Biology and Disease, Columbia Stem Cell Initiative, Columbia University, New York, NY 10032, USA.
| | - Chaolin Zhang
- Departments of Systems Biology and Biochemistry and Molecular Biophysics, Center for Motor Neuron Biology and Disease, Columbia University, New York, NY 10032, USA.
| |
Collapse
|
22
|
Takaku S, Yako H, Niimi N, Akamine T, Kawanami D, Utsunomiya K, Sango K. Establishment of a myelinating co-culture system with a motor neuron-like cell line NSC-34 and an adult rat Schwann cell line IFRS1. Histochem Cell Biol 2018; 149:537-543. [PMID: 29435762 DOI: 10.1007/s00418-018-1649-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/06/2018] [Indexed: 01/01/2023]
Abstract
Co-culture models of neurons and Schwann cells have been utilized for the study of myelination and demyelination in the peripheral nervous system; in most of the previous studies, however, these cells were obtained by primary culture with embryonic or neonatal animals. A spontaneously immortalized Schwann cell line IFRS1 from long-term cultures of adult Fischer rat peripheral nerves has been shown to retain fundamental ability to myelinate neurites in co-cultures with adult rat dorsal root ganglion neurons and nerve growth factor-primed PC12 cells. Our current investigation focuses on the establishment of stable co-culture system with IFRS1 cells and NSC-34 motor neuron-like cells. NSC-34 cells were seeded at a low density (2 × 103/cm2) and maintained for 5-7 days in serum-containing medium supplemented with non-essential amino acids and brain-derived neurotrophic factor (BDNF; 10 ng/mL). Upon observation of neurite outgrowth under a phase-contrast microscope, the NSC-34 cells were exposed to an anti-mitotic agent mitomycin C (1 µg/mL) for 12-16 h, then co-cultured with IFRS1 cells (2 × 104/cm2), and maintained in serum-containing medium supplemented with ascorbic acid (50 µg/mL), BDNF (10 ng/mL), and ciliary neurotrophic factor (10 ng/mL). Double immunofluorescence staining carried out at day 28 of the co-culture showed myelin protein (P0 or PMP22)-immunoreactive IFRS1 cells surrounding the βIII tubulin-immunoreactive neurites. This co-culture system can be a beneficial tool to study the pathogenesis of motor neuron diseases (e.g., amyotrophic lateral sclerosis, Charcot-Marie-Tooth diseases, and immune-mediated demyelinating neuropathies) and novel therapeutic approaches against them.
Collapse
Affiliation(s)
- Shizuka Takaku
- Diabetic Neuropathy Project, Department of Sensory and Motor Systems, Tokyo Metropolitan Institute of Medical Science, 2-1-6 Kamikitazawa, Setagaya-ku, Tokyo, 156-8506, Japan
| | - Hideji Yako
- Diabetic Neuropathy Project, Department of Sensory and Motor Systems, Tokyo Metropolitan Institute of Medical Science, 2-1-6 Kamikitazawa, Setagaya-ku, Tokyo, 156-8506, Japan
| | - Naoko Niimi
- Diabetic Neuropathy Project, Department of Sensory and Motor Systems, Tokyo Metropolitan Institute of Medical Science, 2-1-6 Kamikitazawa, Setagaya-ku, Tokyo, 156-8506, Japan
| | - Tomoyo Akamine
- Diabetic Neuropathy Project, Department of Sensory and Motor Systems, Tokyo Metropolitan Institute of Medical Science, 2-1-6 Kamikitazawa, Setagaya-ku, Tokyo, 156-8506, Japan.,Division of Diabetes, Metabolism and Endocrinology, Department of Internal Medicine, Jikei University School of Medicine, Minato-ku, Tokyo, 105-8461, Japan
| | - Daiji Kawanami
- Division of Diabetes, Metabolism and Endocrinology, Department of Internal Medicine, Jikei University School of Medicine, Minato-ku, Tokyo, 105-8461, Japan
| | - Kazunori Utsunomiya
- Division of Diabetes, Metabolism and Endocrinology, Department of Internal Medicine, Jikei University School of Medicine, Minato-ku, Tokyo, 105-8461, Japan
| | - Kazunori Sango
- Diabetic Neuropathy Project, Department of Sensory and Motor Systems, Tokyo Metropolitan Institute of Medical Science, 2-1-6 Kamikitazawa, Setagaya-ku, Tokyo, 156-8506, Japan.
| |
Collapse
|
23
|
Deriving Dorsal Spinal Sensory Interneurons from Human Pluripotent Stem Cells. Stem Cell Reports 2018; 10:390-405. [PMID: 29337120 PMCID: PMC5832443 DOI: 10.1016/j.stemcr.2017.12.012] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2017] [Revised: 12/14/2017] [Accepted: 12/15/2017] [Indexed: 12/28/2022] Open
Abstract
Cellular replacement therapies for neurological conditions use human embryonic stem cell (hESC)- or induced pluripotent stem cell (hiPSC)-derived neurons to replace damaged or diseased populations of neurons. For the spinal cord, significant progress has been made generating the in-vitro-derived motor neurons required to restore coordinated movement. However, there is as yet no protocol to generate in-vitro-derived sensory interneurons (INs), which permit perception of the environment. Here, we report on the development of a directed differentiation protocol to derive sensory INs for both hESCs and hiPSCs. Two developmentally relevant factors, retinoic acid in combination with bone morphogenetic protein 4, can be used to generate three classes of sensory INs: the proprioceptive dI1s, the dI2s, and mechanosensory dI3s. Critical to this protocol is the competence state of the neural progenitors, which changes over time. This protocol will facilitate developing cellular replacement therapies to reestablish sensory connections in injured patients. Robust protocol to generate spinal sensory neurons from human pluripotent cells RA ± BMP4 direct hPSCs toward the dI1, dI2, and dI3 classes of dorsal interneurons Only neural progenitors in the correct competence state respond to RA/BMP4 signals
Collapse
|
24
|
Cvetkovic C, Rich MH, Raman R, Kong H, Bashir R. A 3D-printed platform for modular neuromuscular motor units. MICROSYSTEMS & NANOENGINEERING 2017; 3:17015. [PMID: 31057862 PMCID: PMC6444989 DOI: 10.1038/micronano.2017.15] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/19/2016] [Revised: 12/23/2016] [Accepted: 01/24/2017] [Indexed: 05/23/2023]
Abstract
A complex and functional living cellular system requires the interaction of one or more cell types to perform specific tasks, such as sensing, processing, or force production. Modular and flexible platforms for fabrication of such multi-cellular modules and their characterization have been lacking. Here, we present a modular cellular system, made up of multi-layered tissue rings containing integrated skeletal muscle and motor neurons (MNs) embedded in an extracellular matrix. The MNs were differentiated from mouse embryonic stem cells through the formation of embryoid bodies (EBs), which are spherical aggregations of cells grown in a suspension culture. The EBs were integrated into a tissue ring with skeletal muscle, which was differentiated in parallel, to create a co-culture amenable to both cell types. The multi-layered rings were then sequentially placed on a stationary three-dimensional-printed hydrogel structure resembling an anatomical muscle-tendon-bone organization. We demonstrate that the site-specific innervation of a group of muscle fibers in the multi-layered tissue rings allows for muscle contraction via chemical stimulation of MNs with glutamate, a major excitatory neurotransmitter in the mammalian nervous system, with the frequency of contraction increasing with glutamate concentration. The addition of tubocurarine chloride (a nicotinic receptor antagonist) halted the contractions, indicating that muscle contraction was MN induced. With a bio-fabricated system permitting controllable mechanical and geometric attributes in a range of length scales, our novel engineered cellular system can be utilized for easier integration of other modular "building blocks" in living cellular and biological machines.
Collapse
Affiliation(s)
- Caroline Cvetkovic
- Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
- Micro and Nanotechnology Laboratory, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Max H. Rich
- Department of Chemical and Biomolecular Engineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Ritu Raman
- Micro and Nanotechnology Laboratory, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
- Department of Mechanical Science and Engineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Hyunjoon Kong
- Department of Chemical and Biomolecular Engineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Rashid Bashir
- Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
- Micro and Nanotechnology Laboratory, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| |
Collapse
|
25
|
Imamura K, Izumi Y, Watanabe A, Tsukita K, Woltjen K, Yamamoto T, Hotta A, Kondo T, Kitaoka S, Ohta A, Tanaka A, Watanabe D, Morita M, Takuma H, Tamaoka A, Kunath T, Wray S, Furuya H, Era T, Makioka K, Okamoto K, Fujisawa T, Nishitoh H, Homma K, Ichijo H, Julien JP, Obata N, Hosokawa M, Akiyama H, Kaneko S, Ayaki T, Ito H, Kaji R, Takahashi R, Yamanaka S, Inoue H. The Src/c-Abl pathway is a potential therapeutic target in amyotrophic lateral sclerosis. Sci Transl Med 2017; 9:eaaf3962. [PMID: 28539470 DOI: 10.1126/scitranslmed.aaf3962] [Citation(s) in RCA: 165] [Impact Index Per Article: 20.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2015] [Revised: 02/04/2016] [Accepted: 12/13/2016] [Indexed: 03/07/2024]
Abstract
Amyotrophic lateral sclerosis (ALS), a fatal disease causing progressive loss of motor neurons, still has no effective treatment. We developed a phenotypic screen to repurpose existing drugs using ALS motor neuron survival as readout. Motor neurons were generated from induced pluripotent stem cells (iPSCs) derived from an ALS patient with a mutation in superoxide dismutase 1 (SOD1). Results of the screen showed that more than half of the hits targeted the Src/c-Abl signaling pathway. Src/c-Abl inhibitors increased survival of ALS iPSC-derived motor neurons in vitro. Knockdown of Src or c-Abl with small interfering RNAs (siRNAs) also rescued ALS motor neuron degeneration. One of the hits, bosutinib, boosted autophagy, reduced the amount of misfolded mutant SOD1 protein, and attenuated altered expression of mitochondrial genes. Bosutinib also increased survival in vitro of ALS iPSC-derived motor neurons from patients with sporadic ALS or other forms of familial ALS caused by mutations in TAR DNA binding protein (TDP-43) or repeat expansions in C9orf72 Furthermore, bosutinib treatment modestly extended survival of a mouse model of ALS with an SOD1 mutation, suggesting that Src/c-Abl may be a potentially useful target for developing new drugs to treat ALS.
Collapse
Affiliation(s)
- Keiko Imamura
- Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto 606-8507, Japan
| | - Yuishin Izumi
- Department of Clinical Neuroscience, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima 770-8503, Japan
| | - Akira Watanabe
- Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto 606-8507, Japan
| | - Kayoko Tsukita
- Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto 606-8507, Japan
| | - Knut Woltjen
- Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto 606-8507, Japan
- Hakubi Center for Advanced Research, Kyoto University, Kyoto 606-8501, Japan
| | - Takuya Yamamoto
- Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto 606-8507, Japan
- Institute for Integrated Cell-Material Sciences, Kyoto University, Kyoto 606-8507, Japan
| | - Akitsu Hotta
- Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto 606-8507, Japan
- Institute for Integrated Cell-Material Sciences, Kyoto University, Kyoto 606-8507, Japan
- Precursory Research for Embryonic Science and Technology (PRESTO), Japan Science and Technology Agency, Saitama 332-0012, Japan
| | - Takayuki Kondo
- Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto 606-8507, Japan
| | - Shiho Kitaoka
- Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto 606-8507, Japan
| | - Akira Ohta
- Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto 606-8507, Japan
| | - Akito Tanaka
- Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto 606-8507, Japan
| | - Dai Watanabe
- Department of Biological Sciences, Graduate School of Medicine, Kyoto University, Kyoto 606-8501, Japan
| | - Mitsuya Morita
- Division of Neurology, Department of Internal Medicine, Jichi Medical University, Tochigi 329-0498, Japan
| | - Hiroshi Takuma
- Department of Neurology, Faculty of Medicine, University of Tsukuba, Tsukuba 305-8575, Japan
| | - Akira Tamaoka
- Department of Neurology, Faculty of Medicine, University of Tsukuba, Tsukuba 305-8575, Japan
| | - Tilo Kunath
- MRC Centre for Regenerative Medicine, School of Biological Sciences, University of Edinburgh, 5 Little France Drive, Edinburgh EH16 4UU, UK
| | - Selina Wray
- Department of Molecular Neuroscience, University College London Institute of Neurology, Queen Square, London WC1N 3BG, UK
| | - Hirokazu Furuya
- Department of Neurology, Kochi Medical School, Kochi University, Kochi 783-8505, Japan
| | - Takumi Era
- Department of Cell Modulation, Institute of Molecular Embryology and Genetics, Kumamoto University, Kumamoto 860-0811, Japan
| | - Kouki Makioka
- Department of Neurology, Gunma University Graduate School of Medicine, Maebashi 371-8511, Japan
| | - Koichi Okamoto
- Geriatrics Research Institute and Hospital, Maebashi 371-0847, Japan
| | - Takao Fujisawa
- Cell Signaling, Graduate School of Pharmaceutical Sciences, University of Tokyo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Hideki Nishitoh
- Department of Medical Sciences, University of Miyazaki, Miyazaki 889-1601, Japan
| | - Kengo Homma
- Cell Signaling, Graduate School of Pharmaceutical Sciences, University of Tokyo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Hidenori Ichijo
- Cell Signaling, Graduate School of Pharmaceutical Sciences, University of Tokyo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Jean-Pierre Julien
- Department of Psychiatry and Neurosciences, Research Centre of Mental Health Institute of Quebec (IUSMQ), Laval University, Québec, Canada
| | - Nanako Obata
- Tokyo Metropolitan Institute of Medical Science, 2-1-6 Kamikitazawa, Setagaya-ku, Tokyo 156-8506, Japan
| | - Masato Hosokawa
- Tokyo Metropolitan Institute of Medical Science, 2-1-6 Kamikitazawa, Setagaya-ku, Tokyo 156-8506, Japan
| | - Haruhiko Akiyama
- Tokyo Metropolitan Institute of Medical Science, 2-1-6 Kamikitazawa, Setagaya-ku, Tokyo 156-8506, Japan
| | - Satoshi Kaneko
- Department of Neurology, Kansai Medical University, Hirakata 573-1191, Japan
| | - Takashi Ayaki
- Department of Neurology, Graduate School of Medicine, Kyoto University, Kyoto 606-8507, Japan
| | - Hidefumi Ito
- Department of Neurology, Wakayama Medical University, Kimiidera, Wakayama 641-8509, Japan
| | - Ryuji Kaji
- Institute for Integrated Cell-Material Sciences, Kyoto University, Kyoto 606-8507, Japan
| | - Ryosuke Takahashi
- Department of Neurology, Graduate School of Medicine, Kyoto University, Kyoto 606-8507, Japan
| | - Shinya Yamanaka
- Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto 606-8507, Japan
- Gladstone Institute of Cardiovascular Disease, San Francisco, CA 94158, USA
| | - Haruhisa Inoue
- Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto 606-8507, Japan.
| |
Collapse
|
26
|
Darvishi M, Tiraihi T, Mesbah-Namin SA, Delshad A, Taheri T. Motor Neuron Transdifferentiation of Neural Stem Cell from Adipose-Derived Stem Cell Characterized by Differential Gene Expression. Cell Mol Neurobiol 2017; 37:275-289. [PMID: 27107758 PMCID: PMC11482063 DOI: 10.1007/s10571-016-0368-x] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2015] [Accepted: 03/24/2016] [Indexed: 02/04/2023]
Abstract
Adipose-derived stem cells (ADSC) are adult stem cells which can be induced into motor neuron-like cells (MNLC) with a preinduction-induction protocol. The purpose of this study is to generate MNLC from neural stem cells (NSC) derived from ADSC. The latter were isolated from the perinephric regions of Sprague-Dawley rats, transdifferentiated into neurospheres (NS) using B27, EGF, and bFGF. After generating NSC from the NS, they induced into MNLC by treating them with Shh and RA, then with GDNF, CNTF, BDNF, and NT-3. The ADSC lineage was evaluated by its mesodermal differentiation and was characterized by immunostaining with CD90, CD105, CD49d, CD106, CD31, CD45, and stemness genes (Oct4, Nanog, and Sox2). The NS and the NSC were evaluated by immunostaining with nestin, NF68, and Neurod1, while the MNLC were evaluated by ISLET1, Olig2, and HB9 genes. The efficiency of MNLC generation was more than 95 ± 1.4 % (mean ± SEM). The in vitro generated myotubes were innervated by the MNLC. The induced ADSC adopted multipolar motor neuron morphology, and they expressed ISLET1, Olig2, and HB9. We conclude that ADSC can be induced into motor neuron phenotype with high efficiency, associated with differential expression of the motor neuron gene. The release of MNLC synaptic vesicles was demonstrated by FM1-43, and they were immunostained with synaptophysin. This activity was correlated with the intracellular calcium ion shift and membrane depolarization upon stimulation as was demonstrated by the calcium indicator and the voltage-sensitive dye, respectively.
Collapse
Affiliation(s)
- Marzieh Darvishi
- Department of Anatomical Sciences, Faculty of Medical Sciences, Tarbiat Modares University, P.O. Box 14155-4838, Tehran, Iran
| | - Taki Tiraihi
- Department of Anatomical Sciences, Faculty of Medical Sciences, Tarbiat Modares University, P.O. Box 14155-4838, Tehran, Iran.
| | - Seyed A Mesbah-Namin
- Department of Clinical Biochemistry, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | | | - Taher Taheri
- Shefa Neurosciences Research Center, Khatam Al-Anbia Hospital, Tehran, Iran
| |
Collapse
|
27
|
Magown P, Rafuse VF, Brownstone RM. Microcircuit formation following transplantation of mouse embryonic stem cell-derived neurons in peripheral nerve. J Neurophysiol 2017; 117:1683-1689. [PMID: 28148646 DOI: 10.1152/jn.00943.2016] [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] [Received: 12/15/2016] [Revised: 01/23/2017] [Accepted: 01/23/2017] [Indexed: 11/22/2022] Open
Abstract
Motoneurons derived from embryonic stem cells can be transplanted in the tibial nerve, where they extend axons to functionally innervate target muscle. Here, we studied spontaneous muscle contractions in these grafts 3 mo following transplantation. One-half of the transplanted grafts generated rhythmic muscle contractions of variable patterns, either spontaneously or in response to brief electrical stimulation. Activity generated by transplanted embryonic stem cell-derived neurons was driven by glutamate and was modulated by muscarinic and GABAergic/glycinergic transmission. Furthermore, rhythmicity was promoted by the same transmitter combination that evokes rhythmic locomotor activity in spinal cord circuits. These results demonstrate that there is a degree of self-assembly of microcircuits in these peripheral grafts involving embryonic stem cell-derived motoneurons and interneurons. Such spontaneous activity is reminiscent of embryonic circuit development in which spontaneous activity is essential for proper connectivity and function and may be necessary for the grafts to form functional connections with muscle.NEW & NOTEWORTHY This manuscript demonstrates that, following peripheral transplantation of neurons derived from embryonic stem cells, the grafts are spontaneously active. The activity is produced and modulated by a number of transmitter systems, indicating that there is a degree of self-assembly of circuits in the grafts.
Collapse
Affiliation(s)
- Philippe Magown
- Medical Neuroscience, Dalhousie University, Halifax, Nova Scotia, Canada.,Department of Surgery (Neurosurgery), Dalhousie University, Halifax, Nova Scotia, Canada
| | - Victor F Rafuse
- Medical Neuroscience, Dalhousie University, Halifax, Nova Scotia, Canada.,Department of Medicine (Neurology), Dalhousie University, Halifax, Nova Scotia, Canada; and
| | - Robert M Brownstone
- Medical Neuroscience, Dalhousie University, Halifax, Nova Scotia, Canada; .,Department of Surgery (Neurosurgery), Dalhousie University, Halifax, Nova Scotia, Canada.,Sobell Department of Motor Neuroscience and Movement Disorders, Institute of Neurology, University College London, London, United Kingdom
| |
Collapse
|
28
|
Wang C, Lu CF, Peng J, Hu CD, Wang Y. Roles of neural stem cells in the repair of peripheral nerve injury. Neural Regen Res 2017; 12:2106-2112. [PMID: 29323053 PMCID: PMC5784362 DOI: 10.4103/1673-5374.221171] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Currently, researchers are using neural stem cell transplantation to promote regeneration after peripheral nerve injury, as neural stem cells play an important role in peripheral nerve injury repair. This article reviews recent research progress of the role of neural stem cells in the repair of peripheral nerve injury. Neural stem cells can not only differentiate into neurons, astrocytes and oligodendrocytes, but can also differentiate into Schwann-like cells, which promote neurite outgrowth around the injury. Transplanted neural stem cells can differentiate into motor neurons that innervate muscles and promote the recovery of neurological function. To promote the repair of peripheral nerve injury, neural stem cells secrete various neurotrophic factors, including brain-derived neurotrophic factor, fibroblast growth factor, nerve growth factor, insulin-like growth factor and hepatocyte growth factor. In addition, neural stem cells also promote regeneration of the axonal myelin sheath, angiogenesis, and immune regulation. It can be concluded that neural stem cells promote the repair of peripheral nerve injury through a variety of ways.
Collapse
Affiliation(s)
- Chong Wang
- Central Hospital of Handan, Handan, Hebei Province; Institute of Orthopedics, Chinese PLA General Hospital, Beijing; Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu Province, China
| | - Chang-Feng Lu
- Institute of Orthopedics, Chinese PLA General Hospital, Beijing, ; Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu Province, China
| | - Jiang Peng
- Institute of Orthopedics, Chinese PLA General Hospital; Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu Province; Beijing Key Lab of Regenerative Medicine in Orthopedics; Key Lab of Musculoskeletal Trauma & War Injuries of Chinese PLA, Beijing, China
| | - Cheng-Dong Hu
- Central Hospital of Handan, Handan, Hebei Province, China
| | - Yu Wang
- Institute of Orthopedics, Chinese PLA General Hospital; Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu Province; Beijing Key Lab of Regenerative Medicine in Orthopedics; Key Lab of Musculoskeletal Trauma & War Injuries of Chinese PLA, Beijing, China
| |
Collapse
|
29
|
Smith AS, Passey SL, Martin NR, Player DJ, Mudera V, Greensmith L, Lewis MP. Creating Interactions between Tissue-Engineered Skeletal Muscle and the Peripheral Nervous System. Cells Tissues Organs 2016; 202:143-158. [PMID: 27825148 PMCID: PMC5175300 DOI: 10.1159/000443634] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/22/2015] [Indexed: 12/22/2022] Open
Abstract
Effective models of mammalian tissues must allow and encourage physiologically (mimetic) correct interactions between co-cultured cell types in order to produce culture microenvironments as similar as possible to those that would normally occur in vivo. In the case of skeletal muscle, the development of such a culture model, integrating multiple relevant cell types within a biomimetic scaffold, would be of significant benefit for investigations into the development, functional performance, and pathophysiology of skeletal muscle tissue. Although some work has been published regarding the behaviour of in vitro muscle models co-cultured with organotypic slices of CNS tissue or with stem cell-derived neurospheres, little investigation has so far been made regarding the potential to maintain isolated motor neurons within a 3D biomimetic skeletal muscle culture platform. Here, we review the current state of the art for engineering neuromuscular contacts in vitro and provide original data detailing the development of a 3D collagen-based model for the co-culture of primary muscle cells and motor neurons. The devised culture system promotes increased myoblast differentiation, forming arrays of parallel, aligned myotubes on which areas of nerve-muscle contact can be detected by immunostaining for pre- and post-synaptic proteins. Quantitative RT-PCR results indicate that motor neuron presence has a positive effect on myotube maturation, suggesting neural incorporation influences muscle development and maturation in vitro. The importance of this work is discussed in relation to other published neuromuscular co-culture platforms along with possible future directions for the field.
Collapse
Affiliation(s)
- Alec S.T. Smith
- Sobell Department of Motor Neuroscience and Movement Disorders, UCL Institute of Neurology, London, UK
- Arthritis Research UK Centre for Sport, Exercise and Osteoarthritis, National Centre for Sport and Exercise Medicine (NCSEM) England, School of Sport, Exercise and Health Sciences, Loughborough University, Loughborough, UK
- Department of Bioengineering, University of Washington, Seattle, Wash., USA
| | - Samantha L. Passey
- Arthritis Research UK Centre for Sport, Exercise and Osteoarthritis, National Centre for Sport and Exercise Medicine (NCSEM) England, School of Sport, Exercise and Health Sciences, Loughborough University, Loughborough, UK
- Department of Pharmacology and Therapeutics, University of Melbourne, Melbourne, Vic., Australia
| | - Neil R.W. Martin
- Arthritis Research UK Centre for Sport, Exercise and Osteoarthritis, National Centre for Sport and Exercise Medicine (NCSEM) England, School of Sport, Exercise and Health Sciences, Loughborough University, Loughborough, UK
| | - Darren J. Player
- Arthritis Research UK Centre for Sport, Exercise and Osteoarthritis, National Centre for Sport and Exercise Medicine (NCSEM) England, School of Sport, Exercise and Health Sciences, Loughborough University, Loughborough, UK
| | - Vivek Mudera
- Division of Surgery and Interventional Science, UCL Institute of Orthopaedics and Musculoskeletal Science, London, UK
| | - Linda Greensmith
- Sobell Department of Motor Neuroscience and Movement Disorders, UCL Institute of Neurology, London, UK
- MRC Centre for Neuromuscular Diseases, UCL Institute of Neurology, London, UK
| | - Mark P. Lewis
- Arthritis Research UK Centre for Sport, Exercise and Osteoarthritis, National Centre for Sport and Exercise Medicine (NCSEM) England, School of Sport, Exercise and Health Sciences, Loughborough University, Loughborough, UK
- *Prof. Mark P. Lewis, School of Sport, Exercise and Health Sciences, Loughborough University, Ashby Road, Loughborough LE11 3TU (UK), E-Mail
| |
Collapse
|
30
|
Liu X, Pfaff DW, Calderon DP, Tabansky I, Wang X, Wang Y, Kow LM. Development of Electrophysiological Properties of Nucleus Gigantocellularis Neurons Correlated with Increased CNS Arousal. Dev Neurosci 2016; 38:295-310. [PMID: 27788521 DOI: 10.1159/000449035] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2016] [Accepted: 08/09/2016] [Indexed: 01/28/2023] Open
Abstract
Many types of data have suggested that neurons in the nucleus gigantocellularis (NGC) in the medullary reticular formation are critically important for CNS arousal and behavioral responsiveness. To extend this topic to a developmental framework, whole-cell patch-recorded characteristics of NGC neurons in brainstem slices and measures of arousal-dependent locomotion of postnatal day 3 (P3) to P6 mouse pups were measured and compared. These neuronal characteristics developed in an orderly, statistically significant monotonic manner over the course of P3-P6: (1) proportion of neurons capable of firing action potential (AP) trains, (2) AP amplitude, (3) AP threshold, (4) amplitude of inward and outward currents, (5) amplitude of negative peak currents, and (6) steady state currents (in I-V plot). These measurements reflect the maturation of sodium and certain potassium channels. Similarly, all measures of locomotion, latency to first movement, total locomotion duration, net locomotion distance, and total quiescence time also developed monotonically over P3-P6. Most importantly, electrophysiological and behavioral measures were significantly correlated. Interestingly, the behavioral measures were not correlated with frequency of excitatory postsynaptic currents or the proportion of neurons showing these currents, responses to a battery of neurotransmitter agents, or rapid activating potassium currents (including IA). Considering the results here in the context of a large body of literature on NGC, we hypothesize that the developmental increase in NGC neuronal excitability participates in causing the increased behavioral responsivity during the postnatal period from P3 to P6.
Collapse
Affiliation(s)
- Xu Liu
- Laboratory of Neurobiology and Behavior, The Rockefeller University, New York, N.Y., USA
| | | | | | | | | | | | | |
Collapse
|
31
|
Cortés D, Robledo-Arratia Y, Hernández-Martínez R, Escobedo-Ávila I, Bargas J, Velasco I. Transgenic GDNF Positively Influences Proliferation, Differentiation, Maturation and Survival of Motor Neurons Produced from Mouse Embryonic Stem Cells. Front Cell Neurosci 2016; 10:217. [PMID: 27672361 PMCID: PMC5018488 DOI: 10.3389/fncel.2016.00217] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2016] [Accepted: 08/30/2016] [Indexed: 11/13/2022] Open
Abstract
Embryonic stem cells (ESC) are pluripotent and thus can differentiate into every cell type present in the body. Directed differentiation into motor neurons (MNs) has been described for pluripotent cells. Although neurotrophic factors promote neuronal survival, their role in neuronal commitment is elusive. Here, we developed double-transgenic lines of mouse ESC (mESC) that constitutively produce glial cell line-derived neurotrophic factor (GDNF) and also contain a GFP reporter, driven by HB9, which is expressed only by postmitotic MNs. After lentiviral transduction, ESC lines integrated and expressed the human GDNF (hGDNF) gene without altering pluripotency markers before differentiation. Further, GDNF-ESC showed significantly higher spontaneous release of this neurotrophin to the medium, when compared to controls. To study MN induction, control and GDNF cell lines were grown as embryoid bodies and stimulated with retinoic acid and Sonic Hedgehog. In GDNF-overexpressing cells, a significant increase of proliferative Olig2+ precursors, which are specified as spinal MNs, was found. Accordingly, GDNF increases the yield of cells with the pan motor neuronal markers HB9, monitored by GFP expression, and Isl1. At terminal differentiation, almost all differentiated neurons express phenotypic markers of MNs in GDNF cultures, with lower proportions in control cells. To test if the effects of GDNF were present at early differentiation stages, exogenous recombinant hGDNF was added to control ESC, also resulting in enhanced MN differentiation. This effect was abolished by the co-addition of neutralizing anti-GDNF antibodies, strongly suggesting that differentiating ESC are responsive to GDNF. Using the HB9::GFP reporter, MNs were selected for electrophysiological recordings. MNs differentiated from GDNF-ESC, compared to control MNs, showed greater electrophysiological maturation, characterized by increased numbers of evoked action potentials (APs), as well as by the appearance of rebound APs, sag inward rectification, spike frequency adaptation and spontaneous synaptic potentials. Upon challenge with kainate, GDNF-overexpressing cells are more resistant to excitotoxicity than control MNs. Together these data indicate that GDNF promotes proliferation of MN-committed precursors, promotes neuronal differentiation, enhances maturation, and confers neuroprotection. GDNF-expressing ESC can be useful in studies of development and disease.
Collapse
Affiliation(s)
- Daniel Cortés
- Instituto de Fisiología Celular-Neurociencias, Universidad Nacional Autónoma de MéxicoMexico City, Mexico; Laboratorio de Reprogramación Celular del Instituto de Fisiología Celular, Universidad Nacional Autónoma de México en el Instituto Nacional de Neurología y Neurología "Manuel Velasco Suárez"Mexico City, Mexico
| | - Yolanda Robledo-Arratia
- Instituto de Fisiología Celular-Neurociencias, Universidad Nacional Autónoma de México Mexico City, Mexico
| | - Ricardo Hernández-Martínez
- Instituto de Fisiología Celular-Neurociencias, Universidad Nacional Autónoma de México Mexico City, Mexico
| | - Itzel Escobedo-Ávila
- Instituto de Fisiología Celular-Neurociencias, Universidad Nacional Autónoma de México Mexico City, Mexico
| | - José Bargas
- Instituto de Fisiología Celular-Neurociencias, Universidad Nacional Autónoma de México Mexico City, Mexico
| | - Iván Velasco
- Instituto de Fisiología Celular-Neurociencias, Universidad Nacional Autónoma de MéxicoMexico City, Mexico; Laboratorio de Reprogramación Celular del Instituto de Fisiología Celular, Universidad Nacional Autónoma de México en el Instituto Nacional de Neurología y Neurología "Manuel Velasco Suárez"Mexico City, Mexico
| |
Collapse
|
32
|
Magown P, Brownstone RM, Rafuse VF. Tumor prevention facilitates delayed transplant of stem cell-derived motoneurons. Ann Clin Transl Neurol 2016; 3:637-49. [PMID: 27606345 PMCID: PMC4999595 DOI: 10.1002/acn3.327] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2016] [Revised: 05/27/2016] [Accepted: 05/31/2016] [Indexed: 12/14/2022] Open
Abstract
OBJECTIVE Nerve injuries resulting in prolonged periods of denervation result in poor recovery of motor function. We have previously shown that embryonic stem cell-derived motoneurons transplanted at the time of transection into a peripheral nerve can functionally reinnervate muscle. For clinical relevance, we now focused on delaying transplantation to assess reinnervation after prolonged denervation. METHODS Embryonic stem cell-derived motoneurons were transplanted into the distal segments of transected tibial nerves in adult mice after prolonged denervation of 1-8 weeks. Twitch and tetanic forces were measured ex vivo 3 months posttransplantation. Tissue was harvested from the transplants for culture and immunohistochemical analysis. RESULTS In this delayed reinnervation model, teratocarcinomas developed in about one half of transplants. A residual multipotent cell population (~ 6% of cells) was found despite neural differentiation. Exposure to the alkylating drug mitomycin C eliminated this multipotent population in vitro while preserving motoneurons. Treating neural differentiated stem cells prior to delayed transplantation prevented tumor formation and resulted in twitch and tetanic forces similar to those in animals transplanted acutely after denervation. INTERPRETATION Despite a neural differentiation protocol, embryonic stem cell-derived motoneurons still carry a risk of tumorigenicity. Pretreating with an antimitotic agent leads to survival and functional muscle reinnervation if performed within 4 weeks of denervation in the mouse.
Collapse
Affiliation(s)
- Philippe Magown
- Medical Neuroscience Dalhousie University Halifax Nova Scotia Canada; Department of Surgery (Neurosurgery) Dalhousie University Halifax Nova Scotia Canada B3H 4R2
| | - Robert M Brownstone
- Medical Neuroscience Dalhousie University Halifax Nova Scotia Canada; Department of Surgery (Neurosurgery) Dalhousie University Halifax Nova Scotia Canada B3H 4R2; Sobell Department of Motor Neuroscience and Movement Disorders Institute of Neurology University College London London WC1N 3BG United Kingdom
| | - Victor F Rafuse
- Medical Neuroscience Dalhousie University Halifax Nova Scotia Canada; Department of Medicine (Neurology) Dalhousie University Halifax Nova Scotia Canada B3H 4R2
| |
Collapse
|
33
|
Uzel SGM, Platt RJ, Subramanian V, Pearl TM, Rowlands CJ, Chan V, Boyer LA, So PTC, Kamm RD. Microfluidic device for the formation of optically excitable, three-dimensional, compartmentalized motor units. SCIENCE ADVANCES 2016; 2:e1501429. [PMID: 27493991 PMCID: PMC4972469 DOI: 10.1126/sciadv.1501429] [Citation(s) in RCA: 148] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/12/2015] [Accepted: 07/06/2016] [Indexed: 05/21/2023]
Abstract
Motor units are the fundamental elements responsible for muscle movement. They are formed by lower motor neurons and their muscle targets, synapsed via neuromuscular junctions (NMJs). The loss of NMJs in neurodegenerative disorders (such as amyotrophic lateral sclerosis or spinal muscle atrophy) or as a result of traumatic injuries affects millions of lives each year. Developing in vitro assays that closely recapitulate the physiology of neuromuscular tissues is crucial to understand the formation and maturation of NMJs, as well as to help unravel the mechanisms leading to their degeneration and repair. We present a microfluidic platform designed to coculture myoblast-derived muscle strips and motor neurons differentiated from mouse embryonic stem cells (ESCs) within a three-dimensional (3D) hydrogel. The device geometry mimics the spinal cord-limb physical separation by compartmentalizing the two cell types, which also facilitates the observation of 3D neurite outgrowth and remote muscle innervation. Moreover, the use of compliant pillars as anchors for muscle strips provides a quantitative functional readout of force generation. Finally, photosensitizing the ESC provides a pool of source cells that can be differentiated into optically excitable motor neurons, allowing for spatiodynamic, versatile, and noninvasive in vitro control of the motor units.
Collapse
Affiliation(s)
- Sebastien G. M. Uzel
- Department of Mechanical Engineering, Massachusetts Institute of Technology (MIT), Cambridge, MA 02139, USA
| | - Randall J. Platt
- Department of Biological Engineering, MIT, Cambridge, MA 02139, USA
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | | | - Taylor M. Pearl
- Department of Biological Engineering, MIT, Cambridge, MA 02139, USA
| | | | - Vincent Chan
- Department of Mechanical Engineering, Massachusetts Institute of Technology (MIT), Cambridge, MA 02139, USA
| | | | - Peter T. C. So
- Department of Mechanical Engineering, Massachusetts Institute of Technology (MIT), Cambridge, MA 02139, USA
- Department of Biological Engineering, MIT, Cambridge, MA 02139, USA
- BioSystems and Micromechanics (BioSyM) IRG, Singapore-MIT Alliance for Research and Technology, Singapore, Singapore
| | - Roger D. Kamm
- Department of Mechanical Engineering, Massachusetts Institute of Technology (MIT), Cambridge, MA 02139, USA
- Department of Biological Engineering, MIT, Cambridge, MA 02139, USA
- BioSystems and Micromechanics (BioSyM) IRG, Singapore-MIT Alliance for Research and Technology, Singapore, Singapore
- Corresponding author.
| |
Collapse
|
34
|
Tadros MA, Fuglevand AJ, Brichta AM, Callister RJ. Intrinsic excitability differs between murine hypoglossal and spinal motoneurons. J Neurophysiol 2016; 115:2672-80. [PMID: 26936988 DOI: 10.1152/jn.01114.2015] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2015] [Accepted: 02/29/2016] [Indexed: 12/12/2022] Open
Abstract
Motoneurons differ in the behaviors they control and their vulnerability to disease and aging. For example, brain stem motoneurons such as hypoglossal motoneurons (HMs) are involved in licking, suckling, swallowing, respiration, and vocalization. In contrast, spinal motoneurons (SMs) innervating the limbs are involved in postural and locomotor tasks requiring higher loads and lower movement velocities. Surprisingly, the properties of these two motoneuron pools have not been directly compared, even though studies on HMs predominate in the literature compared with SMs, especially for adult animals. Here we used whole cell patch-clamp recording to compare the electrophysiological properties of HMs and SMs in age-matched neonatal mice (P7-P10). Passive membrane properties were remarkably similar in HMs and SMs, and afterhyperpolarization properties did not differ markedly between the two populations. HMs had narrower action potentials (APs) and a faster upstroke on their APs compared with SMs. Furthermore, HMs discharged APs at higher frequencies in response to both step and ramp current injection than SMs. Therefore, while HMs and SMs have similar passive properties, they differ in their response to similar levels of depolarizing current. This suggests that each population possesses differing suites of ion channels that allow them to discharge at rates matched to the different mechanical properties of the muscle fibers that drive their distinct motor functions.
Collapse
Affiliation(s)
- M A Tadros
- School of Biomedical Sciences and Pharmacy, Faculty of Health and Hunter Medical Research Institute, The University of Newcastle, Callaghan, New South Wales, Australia; and
| | - A J Fuglevand
- Department of Physiology, College of Medicine, University of Arizona, Tucson, Arizona
| | - A M Brichta
- School of Biomedical Sciences and Pharmacy, Faculty of Health and Hunter Medical Research Institute, The University of Newcastle, Callaghan, New South Wales, Australia; and
| | - R J Callister
- School of Biomedical Sciences and Pharmacy, Faculty of Health and Hunter Medical Research Institute, The University of Newcastle, Callaghan, New South Wales, Australia; and
| |
Collapse
|
35
|
TAKAGI Y. History of Neural Stem Cell Research and Its Clinical Application. Neurol Med Chir (Tokyo) 2016; 56:110-24. [PMID: 26888043 PMCID: PMC4791305 DOI: 10.2176/nmc.ra.2015-0340] [Citation(s) in RCA: 21] [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: 12/18/2015] [Accepted: 01/15/2016] [Indexed: 12/11/2022] Open
Abstract
"Once development was ended…in the adult centers, the nerve paths are something fixed and immutable. Everything may die, nothing may be regenerated," wrote Santiago Ramón y Cajal, a Spanish neuroanatomist and Nobel Prize winner and the father of modern neuroscience. This statement was the central dogma in neuroscience for a long time. However, in the 1960s, neural stem cells (NSCs) were discovered. Since then, our knowledge about NSCs has continued to grow. This review focuses on our current knowledge about NSCs and their surrounding microenvironment. In addition, the clinical application of NSCs for the treatment of various central nervous system diseases is also summarized.
Collapse
Affiliation(s)
- Yasushi TAKAGI
- Department of Neurosurgery, Kyoto University Graduate School of Medicine, Sakyo, Kyoto
| |
Collapse
|
36
|
Retinoic Acid-Mediated Regulation of GLI3 Enables Efficient Motoneuron Derivation from Human ESCs in the Absence of Extrinsic SHH Activation. J Neurosci 2015; 35:11462-81. [PMID: 26290227 DOI: 10.1523/jneurosci.3046-14.2015] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
UNLABELLED The derivation of somatic motoneurons (MNs) from ES cells (ESCs) after exposure to sonic hedgehog (SHH) and retinoic acid (RA) is one of the best defined, directed differentiation strategies to specify fate in pluripotent lineages. In mouse ESCs, MN yield is particularly high after RA + SHH treatment, whereas human ESC (hESC) protocols have been generally less efficient. In an effort to optimize yield, we observe that functional MNs can be derived from hESCs at high efficiencies if treated with patterning molecules at very early differentiation steps before neural induction. Remarkably, under these conditions, equal numbers of human MNs were obtained in the presence or absence of SHH exposure. Using pharmacological and genetic strategies, we demonstrate that early RA treatment directs MN differentiation independently of extrinsic SHH activation by suppressing the induction of GLI3. We further demonstrate that neural induction triggers a switch from a poised to an active chromatin state at GLI3. Early RA treatment prevents this switch by direct binding of the RA receptor at the GLI3 promoter. Furthermore, GLI3 knock-out hESCs can bypass the requirement for early RA patterning to yield MNs efficiently. Our data demonstrate that RA-mediated suppression of GLI3 is sufficient to generate MNs in an SHH-independent manner and that temporal changes in exposure to patterning factors such as RA affect chromatin state and competency of hESC-derived lineages to adopt specific neuronal fates. Finally, our work presents a streamlined platform for the highly efficient derivation of human MNs from ESCs and induced pluripotent stem cells. SIGNIFICANCE STATEMENT Our study presents a rapid and efficient protocol to generate human motoneurons from embryonic and induced pluripotent stem cells. Surprisingly, and in contrast to previous work, motoneurons are generated in the presence of retinoic acid but in the absence of factors that activate sonic hedgehog signaling. We show that early exposure to retinoic acid modulates the chromatin state of cells to be permissive for motoneuron generation and directly suppresses the induction of GLI3, a negative regulator of SHH signaling. Therefore, our data point to a novel mechanism by which retinoic acid exposure can bypass the requirement for extrinsic SHH treatment during motoneuron induction.
Collapse
|
37
|
Zhang Y, Gordon A, Qian W, Chen W. Engineering nanoscale stem cell niche: direct stem cell behavior at cell-matrix interface. Adv Healthc Mater 2015. [PMID: 26222885 DOI: 10.1002/adhm.201500351] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Biophysical cues on the extracellular matrix (ECM) have proven to be significant regulators of stem cell behavior and evolution. Understanding the interplay of these cells and their extracellular microenvironment is critical to future tissue engineering and regenerative medicine, both of which require a means of controlled differentiation. Research suggests that nanotopography, which mimics the local, nanoscale, topographic cues within the stem cell niche, could be a way to achieve large-scale proliferation and control of stem cells in vitro. This Progress Report reviews the history and contemporary advancements of this technology, and pays special attention to nanotopographic fabrication methods and the effect of different nanoscale patterns on stem cell response. Finally, it outlines potential intracellular mechanisms behind this response.
Collapse
Affiliation(s)
- Yan Zhang
- Department of Mechanical and Aerospace Engineering; New York University; Brooklyn NY 11201 USA
| | - Andrew Gordon
- Department of Mechanical and Aerospace Engineering; New York University; Brooklyn NY 11201 USA
| | - Weiyi Qian
- Department of Mechanical and Aerospace Engineering; New York University; Brooklyn NY 11201 USA
| | - Weiqiang Chen
- Department of Mechanical and Aerospace Engineering; New York University; Brooklyn NY 11201 USA
| |
Collapse
|
38
|
Bagher Z, Ebrahimi-Barough S, Azami M, Mirzadeh H, Soleimani M, Ai J, Nourani MR, Joghataei MT. Induction of human umbilical Wharton's jelly-derived mesenchymal stem cells toward motor neuron-like cells. In Vitro Cell Dev Biol Anim 2015; 51:987-94. [PMID: 26148883 DOI: 10.1007/s11626-015-9921-z] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2015] [Accepted: 05/11/2015] [Indexed: 01/06/2023]
Abstract
The most important property of stem cells from different sources is the capacity to differentiate into various cells and tissue types. However, problems including contamination, normal karyotype, and ethical issues cause many limitations in obtaining and using these cells from different sources. The cells in Wharton's jelly region of umbilical cord represent a pool source of primitive cells with properties of mesenchymal stem cells (MSCs). The aim of this study was to determine the potential of human Wharton's jelly-derived mesenchymal stem cells (WJMSCs) for differentiation to motor neuron cells. WJMSCs were induced to differentiate into motor neuron-like cells by using different signaling molecules and neurotrophic factors in vitro. Differentiated neurons were then characterized for expression of motor neuron markers including nestin, PAX6, NF-H, Islet 1, HB9, and choline acetyl transferase (ChAT) by quantitative reverse transcription PCR and immunocytochemistry. Our results showed that differentiated WJMSCs could significantly express motor neuron biomarkers in RNA and protein levels 15 d post induction. These results suggested that WJMSCs can differentiate to motor neuron-like cells and might provide a potential source in cell therapy for neurodegenerative disease.
Collapse
Affiliation(s)
- Zohreh Bagher
- Department of Tissue Engineering and Regenerative Medicine, School of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Somayeh Ebrahimi-Barough
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Mahmoud Azami
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Hamid Mirzadeh
- Polymer Engineering Faculty, Amirkabir University of Technology (Tehran Polytechnic), Tehran, Iran
| | - Mansooreh Soleimani
- Department of Tissue Engineering and Regenerative Medicine, School of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran.,Department of Anatomy, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran.,Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Jafar Ai
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Reza Nourani
- Tissue Engineering Division, Biotechnology Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Mohammad Taghi Joghataei
- Department of Tissue Engineering and Regenerative Medicine, School of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran. .,Department of Anatomy, School of Medicine, Iran University of Medical Science, Tehran, Iran. .,Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
39
|
Park HS, Liu S, McDonald J, Thakor N, Yang IH. Neuromuscular junction in a microfluidic device. ANNUAL INTERNATIONAL CONFERENCE OF THE IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. ANNUAL INTERNATIONAL CONFERENCE 2015; 2013:2833-5. [PMID: 24110317 DOI: 10.1109/embc.2013.6610130] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Malfunctions at the site of neuromuscular junction (NMJ) of post-injuries or diseases are major barriers to recovery of function. The ability to efficiently derive motor neurons (MN) from embryonic stem cells has indicated promise toward the development of new therapies in increasing functional outcomes post injury. Recent advances in micro-technologies have provided advanced culture platforms allowing compartmentalization of sub-cellular components of neurons. In this study, we combined these advances in science and technology to develop a compartmentalized in vitro NMJ model. The developed NMJ system is between mouse embryonic stem cell (mESC)-derived MNs and c2c12 myotubes cultured in a compartmentalized polydimethylsiloxane (PDMS) microfluidic device. While some functional in vitro NMJ systems have been reported, this system would further contribute to research in NMJ-related diseases by providing a system to study the site of action of NMJ aimed at improving promoting better functional recovery.
Collapse
|
40
|
Li Y, Weiss M, Yao L. Directed migration of embryonic stem cell-derived neural cells in an applied electric field. Stem Cell Rev Rep 2015; 10:653-62. [PMID: 24804615 DOI: 10.1007/s12015-014-9518-z] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Spinal cord injury or diseases, such as amyotrophic lateral sclerosis, can cause the loss of motor neurons and therefore results in the paralysis of muscles. Stem cells may improve functional recovery by promoting endogenous regeneration, or by directly replacing neurons. Effective directional migration of grafted neural cells to reconstruct functional connections is crucial in the process. Steady direct current electric fields (EFs) play an important role in the development of the central nervous system. A strong biological effect of EFs is the induction of directional cell migration. In this study, we investigated the guided migration of embryonic stem cell (ESC) derived presumptive motor neurons in an applied EF. The dissociated mouse ESC derived presumptive motor neurons or embryoid bodies were subjected to EFs stimulation and the cell migration was studied. We found that the migration of neural precursors from embryoid bodies was toward cathode pole of applied EFs. Single motor neurons migrated to the cathode of the EFs and reversal of EFs poles reversed their migration direction. The directedness and displacement of cathodal migration became more significant when the field strength was increased from 50 mV/mm to 100 mV/mm. EFs stimulation did not influence the cell migration velocity. Our work suggests that EFs may serve as a guidance cue to direct grafted cell migration in vivo.
Collapse
Affiliation(s)
- Yongchao Li
- Department of Biological Sciences, Wichita State University, Wichita, KS, 67260, USA
| | | | | |
Collapse
|
41
|
Cherry JJ, Kobayashi DT, Lynes MM, Naryshkin NN, Tiziano FD, Zaworski PG, Rubin LL, Jarecki J. Assays for the identification and prioritization of drug candidates for spinal muscular atrophy. Assay Drug Dev Technol 2015; 12:315-41. [PMID: 25147906 DOI: 10.1089/adt.2014.587] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Spinal muscular atrophy (SMA) is an autosomal recessive genetic disorder resulting in degeneration of α-motor neurons of the anterior horn and proximal muscle weakness. It is the leading cause of genetic mortality in children younger than 2 years. It affects ∼1 in 11,000 live births. In 95% of cases, SMA is caused by homozygous deletion of the SMN1 gene. In addition, all patients possess at least one copy of an almost identical gene called SMN2. A single point mutation in exon 7 of the SMN2 gene results in the production of low levels of full-length survival of motor neuron (SMN) protein at amounts insufficient to compensate for the loss of the SMN1 gene. Although no drug treatments are available for SMA, a number of drug discovery and development programs are ongoing, with several currently in clinical trials. This review describes the assays used to identify candidate drugs for SMA that modulate SMN2 gene expression by various means. Specifically, it discusses the use of high-throughput screening to identify candidate molecules from primary screens, as well as the technical aspects of a number of widely used secondary assays to assess SMN messenger ribonucleic acid (mRNA) and protein expression, localization, and function. Finally, it describes the process of iterative drug optimization utilized during preclinical SMA drug development to identify clinical candidates for testing in human clinical trials.
Collapse
|
42
|
Motoneurons derived from induced pluripotent stem cells develop mature phenotypes typical of endogenous spinal motoneurons. J Neurosci 2015; 35:1291-306. [PMID: 25609642 DOI: 10.1523/jneurosci.2126-14.2015] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Induced pluripotent cell-derived motoneurons (iPSCMNs) are sought for use in cell replacement therapies and treatment strategies for motoneuron diseases such as amyotrophic lateral sclerosis (ALS). However, much remains unknown about the physiological properties of iPSCMNs and how they compare with endogenous spinal motoneurons or embryonic stem cell-derived motoneurons (ESCMNs). In the present study, we first used a proteomic approach and compared protein expression profiles between iPSCMNs and ESCMNs to show that <4% of the proteins identified were differentially regulated. Like ESCs, we found that mouse iPSCs treated with retinoic acid and a smoothened agonist differentiated into motoneurons expressing the LIM homeodomain protein Lhx3. When transplanted into the neural tube of developing chick embryos, iPSCMNs selectively targeted muscles normally innervated by Lhx3 motoneurons. In vitro studies showed that iPSCMNs form anatomically mature and functional neuromuscular junctions (NMJs) when cocultured with chick myofibers for several weeks. Electrophysiologically, iPSCMNs developed passive membrane and firing characteristic typical of postnatal motoneurons after several weeks in culture. Finally, iPSCMNs grafted into transected mouse tibial nerve projected axons to denervated gastrocnemius muscle fibers, where they formed functional NMJs, restored contractile force. and attenuated denervation atrophy. Together, iPSCMNs possess many of the same cellular and physiological characteristics as ESCMNs and endogenous spinal motoneurons. These results further justify using iPSCMNs as a source of motoneurons for cell replacement therapies and to study motoneuron diseases such as ALS.
Collapse
|
43
|
Smith AST, Long CJ, McAleer C, Guo X, Esch M, Prot JM, Shuler ML, Hickman JJ. ‘Body-on-a-Chip’ Technology and Supporting Microfluidics. HUMAN-BASED SYSTEMS FOR TRANSLATIONAL RESEARCH 2014. [DOI: 10.1039/9781782620136-00132] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
In order to effectively streamline current drug development protocols, there is a need to generate high information content preclinical screens capable of generating data with a predictive power in relation to the activity of novel therapeutics in humans. Given the poor predictive power of animal models, and the lack of complexity and interconnectivity of standard in vitro culture methodologies, many investigators are now moving toward the development of physiologically and functionally accurate culture platforms composed of human cells to investigate cellular responses to drug compounds in high-throughput preclinical studies. The generation of complex, multi-organ in vitro platforms, built to recapitulate physiological dimensions, flow rates and shear stresses, is being investigated as the logical extension of this drive. Production and application of a biologically accurate multi-organ platform, or ‘body-on-a-chip’, would facilitate the correct modelling of the dynamic and interconnected state of living systems for high-throughput drug studies as well as basic and applied biomolecular research. This chapter will discuss current technologies aimed at producing ‘body-on-a-chip’ models, as well as highlighting recent advances and important challenges still to be met in the development of biomimetic single-organ systems for drug development purposes.
Collapse
Affiliation(s)
- A. S. T. Smith
- NanoScience Technology Center, University of Central Florida Orlando FL 32826 USA
| | - C. J. Long
- NanoScience Technology Center, University of Central Florida Orlando FL 32826 USA
| | - C. McAleer
- NanoScience Technology Center, University of Central Florida Orlando FL 32826 USA
| | - X. Guo
- NanoScience Technology Center, University of Central Florida Orlando FL 32826 USA
| | - M. Esch
- Biomedical Engineering, Cornell University Ithaca NY USA
| | - J. M. Prot
- Biomedical Engineering, Cornell University Ithaca NY USA
| | - M. L. Shuler
- Biomedical Engineering, Cornell University Ithaca NY USA
| | - J. J. Hickman
- NanoScience Technology Center, University of Central Florida Orlando FL 32826 USA
| |
Collapse
|
44
|
Maury Y, Côme J, Piskorowski RA, Salah-Mohellibi N, Chevaleyre V, Peschanski M, Martinat C, Nedelec S. Combinatorial analysis of developmental cues efficiently converts human pluripotent stem cells into multiple neuronal subtypes. Nat Biotechnol 2014; 33:89-96. [PMID: 25383599 DOI: 10.1038/nbt.3049] [Citation(s) in RCA: 267] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2014] [Accepted: 09/19/2014] [Indexed: 12/19/2022]
Abstract
Specification of cell identity during development depends on exposure of cells to sequences of extrinsic cues delivered at precise times and concentrations. Identification of combinations of patterning molecules that control cell fate is essential for the effective use of human pluripotent stem cells (hPSCs) for basic and translational studies. Here we describe a scalable, automated approach to systematically test the combinatorial actions of small molecules for the targeted differentiation of hPSCs. Applied to the generation of neuronal subtypes, this analysis revealed an unappreciated role for canonical Wnt signaling in specifying motor neuron diversity from hPSCs and allowed us to define rapid (14 days), efficient procedures to generate spinal and cranial motor neurons as well as spinal interneurons and sensory neurons. Our systematic approach to improving hPSC-targeted differentiation should facilitate disease modeling studies and drug screening assays.
Collapse
Affiliation(s)
- Yves Maury
- CECS, I-STEM (Institute for Stem Cell Therapy and Exploration of Monogenic Diseases), AFM, Evry, France
| | - Julien Côme
- CECS, I-STEM (Institute for Stem Cell Therapy and Exploration of Monogenic Diseases), AFM, Evry, France
| | | | | | - Vivien Chevaleyre
- CNRS UMR 8118, Université Paris Descartes Sorbonne Paris Cité, Paris, France
| | | | | | | |
Collapse
|
45
|
Montgomery A, Wong A, Gabers N, Willerth SM. Engineering personalized neural tissue by combining induced pluripotent stem cells with fibrin scaffolds. Biomater Sci 2014. [PMID: 26218131 DOI: 10.1039/c4bm00299g] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Induced pluripotent stem cells (iPSCs) are generated from adult somatic cells through the induction of key transcription factors that restore the ability to become any cell type found in the body. These cells are of interest for tissue engineering due to their potential for developing patient-specific therapies. As the technology for generating iPSCs advances, it is important to concurrently investigate protocols for the efficient differentiation of these cells to desired downstream phenotypes in combination with biomaterial scaffolds as a way of engineering neural tissue. For such applications, the generation of neurons within three dimensional fibrin scaffolds has been well characterized as a cell-delivery platform for murine embryonic stem cells (ESCs) but has not yet been applied to murine iPSCs. Given that iPSCs have been reported to differentiate less effectively than ESCs, a key objective of this investigation is to maximize the proportion of iPSC-derived neurons in fibrin through the choice of differentiation protocol. To this end, this study compares two EB-mediated protocols for generating neurons from murine iPSCs and ESCs: an 8 day 4-/4+ protocol using soluble retinoic acid in the last 4 days and a 6 day 2-/4+ protocol using soluble retinoic acid and the small molecule sonic hedgehog agonist purmorphamine in the last 4 days. EBs were then seeded in fibrin scaffolds for 14 days to allow further differentiation into neurons. EBs generated by the 2-/4+ protocol yielded a higher percentage of neurons compared to those from the 4-/4+ protocol for both iPSCs and ESCs. The results demonstrate the successful translation of the fibrin-based cell-delivery platform for use with murine iPSCs and furthermore that the proportion of neurons generated from murine iPSC-derived EBs seeded in fibrin can be maximized using the 2-/4+ differentiation protocol. Together, these findings validate the further exploration of 3D fibrin-based scaffolds as a method of delivering neuronal cells derived from iPSCs - an important step toward the development of iPSC-based tissue engineering strategies for spinal cord injury repair.
Collapse
Affiliation(s)
- Amy Montgomery
- Department of Mechanical Engineering, University of Victoria, Canada.
| | | | | | | |
Collapse
|
46
|
Abdanipour A, Tiraihi T, Taheri T. Intraspinal transplantation of motoneuron-like cell combined with delivery of polymer-based glial cell line-derived neurotrophic factor for repair of spinal cord contusion injury. Neural Regen Res 2014; 9:1003-13. [PMID: 25206752 PMCID: PMC4146307 DOI: 10.4103/1673-5374.133159] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/18/2014] [Indexed: 12/28/2022] Open
Abstract
To evaluate the effects of glial cell line-derived neurotrophic factor transplantation combined with adipose-derived stem cells-transdifferentiated motoneuron delivery on spinal cord contusion injury, we developed rat models of spinal cord contusion injury, 7 days later, injected adipose-derived stem cells-transdifferentiated motoneurons into the epicenter, rostral and caudal regions of the impact site and simultaneously transplanted glial cell line-derived neurotrophic factor-gelfoam complex into the myelin sheath. Motoneuron-like cell transplantation combined with glial cell line-derived neurotrophic factor delivery reduced cavity formations and increased cell density in the transplantation site. The combined therapy exhibited superior promoting effects on recovery of motor function to transplantation of glial cell line-derived neurotrophic factor, adipose-derived stem cells or motoneurons alone. These findings suggest that motoneuron-like cell transplantation combined with glial cell line-derived neurotrophic factor delivery holds a great promise for repair of spinal cord injury.
Collapse
Affiliation(s)
- Alireza Abdanipour
- Shefa Neuroscience Research Center at Khatam Al-Anbia Hospital, Tehran, Iran
| | - Taki Tiraihi
- Shefa Neuroscience Research Center at Khatam Al-Anbia Hospital, Tehran, Iran
| | - Taher Taheri
- Shefa Neuroscience Research Center at Khatam Al-Anbia Hospital, Tehran, Iran
| |
Collapse
|
47
|
Maeda M, Harris AW, Kingham BF, Lumpkin CJ, Opdenaker LM, McCahan SM, Wang W, Butchbach MER. Transcriptome profiling of spinal muscular atrophy motor neurons derived from mouse embryonic stem cells. PLoS One 2014; 9:e106818. [PMID: 25191843 PMCID: PMC4156416 DOI: 10.1371/journal.pone.0106818] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2014] [Accepted: 08/01/2014] [Indexed: 01/20/2023] Open
Abstract
Proximal spinal muscular atrophy (SMA) is an early onset, autosomal recessive motor neuron disease caused by loss of or mutation in SMN1 (survival motor neuron 1). Despite understanding the genetic basis underlying this disease, it is still not known why motor neurons (MNs) are selectively affected by the loss of the ubiquitously expressed SMN protein. Using a mouse embryonic stem cell (mESC) model for severe SMA, the RNA transcript profiles (transcriptomes) between control and severe SMA (SMN2+/+;mSmn−/−) mESC-derived MNs were compared in this study using massively parallel RNA sequencing (RNA-Seq). The MN differentiation efficiencies between control and severe SMA mESCs were similar. RNA-Seq analysis identified 3,094 upregulated and 6,964 downregulated transcripts in SMA mESC-derived MNs when compared against control cells. Pathway and network analysis of the differentially expressed RNA transcripts showed that pluripotency and cell proliferation transcripts were significantly increased in SMA MNs while transcripts related to neuronal development and activity were reduced. The differential expression of selected transcripts such as Crabp1, Crabp2 and Nkx2.2 was validated in a second mESC model for SMA as well as in the spinal cords of low copy SMN2 severe SMA mice. Furthermore, the levels of these selected transcripts were restored in high copy SMN2 rescue mouse spinal cords when compared against low copy SMN2 severe SMA mice. These findings suggest that SMN deficiency affects processes critical for normal development and maintenance of MNs.
Collapse
Affiliation(s)
- Miho Maeda
- Center for Applied Clinical Genomics, Nemours Biomedical Research, Nemours Alfred I. duPont Hospital for Children, Wilmington, Delaware, United States of America
- Department of Biological Sciences, University of Delaware, Newark, Delaware, United States of America
| | - Ashlee W. Harris
- Center for Applied Clinical Genomics, Nemours Biomedical Research, Nemours Alfred I. duPont Hospital for Children, Wilmington, Delaware, United States of America
| | - Brewster F. Kingham
- Sequencing and Genotyping Center, University of Delaware, Newark, Delaware, United States of America
| | - Casey J. Lumpkin
- Center for Applied Clinical Genomics, Nemours Biomedical Research, Nemours Alfred I. duPont Hospital for Children, Wilmington, Delaware, United States of America
- Department of Biological Sciences, University of Delaware, Newark, Delaware, United States of America
| | - Lynn M. Opdenaker
- Center for Translational Cancer Research, University of Delaware, Newark, Delaware, United States of America
| | - Suzanne M. McCahan
- Center for Pediatric Research, Nemours Biomedical Research, Nemours Alfred I. duPont Hospital for Children, Wilmington, Delaware, United States of America
- Bioinformatics Core Facility, Nemours Biomedical Research, Nemours Alfred I. duPont Hospital for Children, Wilmington, Delaware, United States of America
- Department of Pediatrics, Thomas Jefferson University, Philadelphia, Pennsylvania, United States of America
| | - Wenlan Wang
- Center for Applied Clinical Genomics, Nemours Biomedical Research, Nemours Alfred I. duPont Hospital for Children, Wilmington, Delaware, United States of America
- Center for Pediatric Research, Nemours Biomedical Research, Nemours Alfred I. duPont Hospital for Children, Wilmington, Delaware, United States of America
- Department of Biological Sciences, University of Delaware, Newark, Delaware, United States of America
| | - Matthew E. R. Butchbach
- Center for Applied Clinical Genomics, Nemours Biomedical Research, Nemours Alfred I. duPont Hospital for Children, Wilmington, Delaware, United States of America
- Center for Pediatric Research, Nemours Biomedical Research, Nemours Alfred I. duPont Hospital for Children, Wilmington, Delaware, United States of America
- Department of Biological Sciences, University of Delaware, Newark, Delaware, United States of America
- Department of Pediatrics, Thomas Jefferson University, Philadelphia, Pennsylvania, United States of America
- * E-mail:
| |
Collapse
|
48
|
Uzel SGM, Pavesi A, Kamm RD. Microfabrication and microfluidics for muscle tissue models. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2014; 115:279-93. [PMID: 25175338 DOI: 10.1016/j.pbiomolbio.2014.08.013] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/16/2014] [Accepted: 08/19/2014] [Indexed: 12/14/2022]
Abstract
The relatively recent development of microfluidic systems with wide-ranging capabilities for generating realistic 2D or 3D systems with single or multiple cell types has given rise to an extensive collection of platform technologies useful in muscle tissue engineering. These new systems are aimed at (i) gaining fundamental understanding of muscle function, (ii) creating functional muscle constructs in vitro, and (iii) utilizing these constructs a variety of applications. Use of microfluidics to control the various stimuli that promote differentiation of multipotent cells into cardiac or skeletal muscle is first discussed. Next, systems that incorporate muscle cells to produce either 2D sheets or 3D tissues of contractile muscle are described with an emphasis on the more recent 3D platforms. These systems are useful for fundamental studies of muscle biology and can also be incorporated into drug screening assays. Applications are discussed for muscle actuators in the context of microrobotics and in miniaturized biological pumps. Finally, an important area of recent study involves coculture with cell types that either activate muscle or facilitate its function. Limitations of current designs and the potential for improving functionality for a wider range of applications is also discussed, with a look toward using current understanding and capabilities to design systems of greater realism, complexity and functionality.
Collapse
Affiliation(s)
- Sebastien G M Uzel
- Department of Mechanical Engineering, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA 02139, USA
| | - Andrea Pavesi
- Singapore MIT Alliance for Research and Technology, BioSystems and Micromechanics, 1 CREATE way, #04-13/14 Enterprise Wing, Singapore 138602, Singapore
| | - Roger D Kamm
- Department of Mechanical Engineering, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA 02139, USA; Singapore MIT Alliance for Research and Technology, BioSystems and Micromechanics, 1 CREATE way, #04-13/14 Enterprise Wing, Singapore 138602, Singapore; Department of Biological Engineering, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA 02139, USA.
| |
Collapse
|
49
|
Lu D, Chen EYT, Lee P, Wang YC, Ching W, Markey C, Gulstrom C, Chen LC, Nguyen T, Chin WC. Accelerated neuronal differentiation toward motor neuron lineage from human embryonic stem cell line (H9). Tissue Eng Part C Methods 2014; 21:242-52. [PMID: 25036750 DOI: 10.1089/ten.tec.2013.0725] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Motor neurons loss plays a pivotal role in the pathoetiology of various debilitating diseases such as, but not limited to, amyotrophic lateral sclerosis, primary lateral sclerosis, progressive muscular atrophy, progressive bulbar palsy, pseudobulbar palsy, and spinal muscular atrophy. However, advancement in motor neuron replacement therapy has been significantly constrained by the difficulties in large-scale production at a cost-effective manner. Current methods to derive motor neuron heavily rely on biochemical stimulation, chemical biological screening, and complex physical cues. These existing methods are seriously challenged by extensive time requirements and poor yields. An innovative approach that overcomes prior hurdles and enhances the rate of successful motor neuron transplantation in patients is of critical demand. Iron, a trace element, is indispensable for the normal development and function of the central nervous system. Whether ferric ions promote neuronal differentiation and subsequently promote motor neuron lineage has never been considered. Here, we demonstrate that elevated iron concentration can drastically accelerate the differentiation of human embryonic stem cells (hESCs) toward motor neuron lineage potentially via a transferrin mediated pathway. HB9 expression in 500 nM iron-treated hESCs is approximately twofold higher than the control. Moreover, iron treatment generated more matured and functional motor neuron-like cells that are ∼1.5 times more sensitive to depolarization when compared to the control. Our methodology renders an expedited approach to harvest motor neuron-like cells for disease, traumatic injury regeneration, and drug screening.
Collapse
Affiliation(s)
- David Lu
- 1 Bioengineering Program, School of Engineering, University of California , Merced, California
| | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
McKinnon DD, Brown TE, Kyburz KA, Kiyotake E, Anseth KS. Design and characterization of a synthetically accessible, photodegradable hydrogel for user-directed formation of neural networks. Biomacromolecules 2014; 15:2808-16. [PMID: 24932668 PMCID: PMC4592536 DOI: 10.1021/bm500731b] [Citation(s) in RCA: 75] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Hydrogels with photocleavable units incorporated into the cross-links have provided researchers with the ability to control mechanical properties temporally and study the role of matrix signaling on stem cell function and fate. With a growing interest in dynamically tunable cell culture systems, methods to synthesize photolabile hydrogels from simple precursors would facilitate broader accessibility. Here, a step-growth photodegradable poly(ethylene glycol) (PEG) hydrogel system cross-linked through a strain promoted alkyne-azide cycloaddition (SPAAC) reaction and degraded through the cleavage of a nitrobenzyl ether moiety integrated into the cross-links is developed from commercially available precursors in three straightforward synthetic steps with high yields (>95%). The network evolution and degradation properties are characterized in response to one- and two-photon irradiation. The PEG hydrogel is employed to encapsulate embryonic stem cell-derived motor neurons (ESMNs), and in situ degradation is exploited to gain three-dimensional control over the extension of motor axons using two-photon infrared light. Finally, ESMNs and their in vivo synaptic partners, myotubes, are coencapsulated, and the formation of user-directed neural networks is demonstrated.
Collapse
Affiliation(s)
- Daniel D. McKinnon
- Department of Chemical and Biological Engineering, University of Colorado, Boulder, Colorado 80303, United States
- Howard Hughes Medical Institute, University of Colorado, Boulder, Colorado 80303, United States
| | - Tobin E. Brown
- Department of Chemical and Biological Engineering, University of Colorado, Boulder, Colorado 80303, United States
- Howard Hughes Medical Institute, University of Colorado, Boulder, Colorado 80303, United States
| | - Kyle A. Kyburz
- Department of Chemical and Biological Engineering, University of Colorado, Boulder, Colorado 80303, United States
- Howard Hughes Medical Institute, University of Colorado, Boulder, Colorado 80303, United States
| | - Emi Kiyotake
- Department of Chemical and Biological Engineering, University of Colorado, Boulder, Colorado 80303, United States
- Howard Hughes Medical Institute, University of Colorado, Boulder, Colorado 80303, United States
| | - Kristi S. Anseth
- Department of Chemical and Biological Engineering, University of Colorado, Boulder, Colorado 80303, United States
- BioFrontiers Institute, University of Colorado, Boulder, Colorado 80303, United States
- Howard Hughes Medical Institute, University of Colorado, Boulder, Colorado 80303, United States
| |
Collapse
|