1
|
Yang Y, Akdemir AR, Rashik RA, Shihadeh Khater OA, Weng Z, Wang L, Zhong Y, Gallant ND. Guided neural stem cell differentiation by dynamic loading of 3D printed elastomeric scaffolds. J Mech Behav Biomed Mater 2025; 165:106940. [PMID: 39955829 DOI: 10.1016/j.jmbbm.2025.106940] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Revised: 01/22/2025] [Accepted: 02/09/2025] [Indexed: 02/18/2025]
Abstract
The limited regenerative ability of "permanent" cells is a major barrier to treating conditions like spinal cord injury (SCI) and myocardial infarction (MI). The delivery of stem cells, which can generate various cell types, offer potential for personalized therapy with reduced immunoreaction and recovery time. However, restoring function to these tissues also requires new or replacement cells to align properly. Neurons, for example, must organize and extend parallel axons, mimicking their natural structure for directional signal propagation. Current stem cell differentiation methods lack guidance, resulting in randomly distributed axons and limited repair effectiveness. Advancing methods and materials to guide stem cell differentiation into functional, aligned nerve bundles is crucial for improving SCI treatment outcomes. This study aimed to develop an in vitro system to promote aligned neural differentiation by applying cyclic uniaxial tension to PC-12 stem cells adhered to 3D-printed elastic scaffolds. We created a simple loading device which can apply cyclic and controllable stretching force to a scaffold, which in turn transmits uniaxial tension to cells adhered to the scaffold during their differentiation. An elastomer ink for 3D printing scaffolds was formulated and surface treatment processes were investigated to enhance the cell-scaffold adhesion to support the dynamic loading. It was revealed that a corona discharge treatment while the scaffold is soaked with type I collagen can significantly enhance cell adhesion. A range of strain magnitudes and frequencies were revealed to enhance the differentiation of neural tissue derived PC-12 cells to neuron cells and increase the length of their neurites up to 76%. The combination of 3% maximum strain and 1 Hz loading frequency maximized differentiation and neurite extension. These findings demonstrate that dynamic mechanical stimulation enhances neural differentiation and organization, offering an alternative approach for regenerative therapies targeting SCI and similar conditions.
Collapse
Affiliation(s)
- Yi Yang
- Department of Mechanical Engineering, University of South Florida, 4202 E. Fowler Ave, Tampa, FL, 33620, USA
| | - Abdullah Revaha Akdemir
- Department of Mechanical Engineering, University of South Florida, 4202 E. Fowler Ave, Tampa, FL, 33620, USA
| | - Rafsan Ahmed Rashik
- Department of Mechanical Engineering, University of South Florida, 4202 E. Fowler Ave, Tampa, FL, 33620, USA
| | - Omar Ahmad Shihadeh Khater
- Department of Mechanical Engineering, University of South Florida, 4202 E. Fowler Ave, Tampa, FL, 33620, USA
| | - Zijian Weng
- Department of Mechanical Engineering, University of South Florida, 4202 E. Fowler Ave, Tampa, FL, 33620, USA
| | - Long Wang
- Department of Civil and Environmental Engineering, California Polytechnic State University, San Luis Obispo, CA, 93407, USA
| | - Ying Zhong
- Sauvage Laboratory for Smart Materials, School of Integrated Circuits, Harbin Institute of Technology (Shenzhen), University Town of Shenzhen, Shenzhen, Guangdong, 518055, China.
| | - Nathan D Gallant
- Department of Mechanical Engineering, University of South Florida, 4202 E. Fowler Ave, Tampa, FL, 33620, USA.
| |
Collapse
|
2
|
Wang NB, Adewumi HO, Lende-Dorn BA, Beitz AM, O'Shea TM, Galloway KE. Compact transcription factor cassettes generate functional, engraftable motor neurons by direct conversion. Cell Syst 2025; 16:101206. [PMID: 40086435 DOI: 10.1016/j.cels.2025.101206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Revised: 11/07/2024] [Accepted: 02/11/2025] [Indexed: 03/16/2025]
Abstract
Direct conversion generates patient-specific, disease-relevant cell types, such as neurons, that are rare, limited, or difficult to isolate from common and easily accessible cells, such as skin cells. However, low rates of direct conversion and complex protocols limit scalability and, thus, the potential of cell-fate conversion for biomedical applications. Here, we optimize the conversion protocol by examining process parameters, including transcript design; delivery via adeno-associated virus (AAV), retrovirus, and lentivirus; cell seeding density; and the impact of media conditions. Thus, we report a compact, portable conversion process that boosts proliferation and increases direct conversion of mouse fibroblasts to induced motor neurons (iMNs) to achieve high conversion rates of above 1,000%, corresponding to more than ten motor neurons yielded per cell seeded, which we achieve through expansion. Our optimized, direct conversion process generates functional motor neurons at scales relevant for cell therapies (>107 cells) that graft with the mouse central nervous system. High-efficiency, compact, direct conversion systems will support scaling to patient-specific, neural cell therapies.
Collapse
Affiliation(s)
- Nathan B Wang
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Honour O Adewumi
- Department of Biomedical Engineering, Boston University, Boston, MA 02215, USA
| | - Brittany A Lende-Dorn
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Adam M Beitz
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Timothy M O'Shea
- Department of Biomedical Engineering, Boston University, Boston, MA 02215, USA
| | - Kate E Galloway
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA.
| |
Collapse
|
3
|
Sun P, Yuan Y, Lv Z, Yu X, Ma H, Liang S, Zhang J, Zhu J, Lu J, Wang C, Huan L, Jin C, Wang C, Li W. Generation of self-renewing neuromesodermal progenitors with neuronal and skeletal muscle bipotential from human embryonic stem cells. CELL REPORTS METHODS 2024; 4:100897. [PMID: 39515335 PMCID: PMC11705767 DOI: 10.1016/j.crmeth.2024.100897] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Revised: 07/19/2024] [Accepted: 10/15/2024] [Indexed: 11/16/2024]
Abstract
Progress has been made in generating spinal cord and trunk derivatives from neuromesodermal progenitors (NMPs). However, maintaining the self-renewal of NMPs in vitro remains a challenge. In this study, we developed a cocktail of small molecules and growth factors that induces human embryonic stem cells to produce self-renewing NMPs (srNMPs) under chemically defined conditions. These srNMPs maintain the state of neuromesodermal progenitors in prolonged culture and have the potential to generate mesodermal cells and neurons, even at the single-cell level. Additionally, suspended srNMP aggregates can spontaneously differentiate into all tissue types of early embryonic trunks. Furthermore, transplanted srNMP-derived muscle satellite cells or progenitors of motor neurons were integrated into skeletal muscle or the spinal cord, respectively, and contributed to regeneration in mouse models. In summary, srNMPs hold great promise for applications in developmental biology and as renewable cell sources for cell therapy for trunk and spinal cord injuries.
Collapse
Affiliation(s)
- Pingxin Sun
- Department of Cell Biology, Naval Medical University, 200433 Shanghai, China
| | - Yuan Yuan
- Department of Cell Biology, Naval Medical University, 200433 Shanghai, China
| | - Zhuman Lv
- Department of Cell Biology, Naval Medical University, 200433 Shanghai, China
| | - Xinlu Yu
- Department of Cell Biology, Naval Medical University, 200433 Shanghai, China
| | - Haoxin Ma
- Department of Cell Biology, Naval Medical University, 200433 Shanghai, China
| | - Shulong Liang
- Department of Cell Biology, Naval Medical University, 200433 Shanghai, China
| | - Jiqianzhu Zhang
- Department of Cell Biology, Naval Medical University, 200433 Shanghai, China; Department of Health Toxicology, Naval Medical University, 200433 Shanghai, China
| | - Jiangbo Zhu
- Department of Health Toxicology, Naval Medical University, 200433 Shanghai, China
| | - Junyu Lu
- Department of Cell Biology, Naval Medical University, 200433 Shanghai, China
| | - Chunyan Wang
- Department of Cell Biology, Naval Medical University, 200433 Shanghai, China
| | - Le Huan
- Department of Cell Biology, Naval Medical University, 200433 Shanghai, China.
| | - Caixia Jin
- Department of Regenerative Medicine, College of Medicine, Tongji University, 200433 Shanghai, China.
| | - Chao Wang
- Department of Cell Biology, Naval Medical University, 200433 Shanghai, China.
| | - Wenlin Li
- Department of Cell Biology, Naval Medical University, 200433 Shanghai, China; Shanghai Key Laboratory of Cell Engineering, Naval Medical University, 200433 Shanghai, China.
| |
Collapse
|
4
|
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
|
5
|
Rabeling A, van der Hoven A, Andersen N, Goolam M. Neural Tube Organoids: A Novel System to Study Developmental Timing. Stem Cell Rev Rep 2024; 20:2045-2061. [PMID: 39230820 PMCID: PMC11554929 DOI: 10.1007/s12015-024-10785-5] [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] [Accepted: 08/27/2024] [Indexed: 09/05/2024]
Abstract
The neural tube (NT) is a transient structure formed during embryogenesis which develops into the brain and spinal cord. While mouse models have been commonly used in place of human embryos to study NT development, species-specific differences limit their applicability. One major difference is developmental timing, with NT formation from the neural plate in 16 days in humans compared to 4 days in mice, as well as differences in the time taken to form neuronal subtypes and complete neurogenesis. Neural tube organoids (NTOs) represent a new way to study NT development in vitro. While mouse and human NTOs have been shown to recapitulate the major developmental events of NT formation; it is unknown whether species-specific developmental timing, also termed allochrony, is also recapitulated. This review summarises current research using both mouse and human NTOs and compares developmental timing events in order to assess if allochrony is maintained in organoids.
Collapse
Affiliation(s)
- Alexa Rabeling
- Department of Human Biology, Faculty of Health Sciences, University of Cape Town, Cape Town, 7925, South Africa
- UCT Neuroscience Institute, Cape Town, South Africa
| | - Amy van der Hoven
- Department of Human Biology, Faculty of Health Sciences, University of Cape Town, Cape Town, 7925, South Africa
- UCT Neuroscience Institute, Cape Town, South Africa
| | - Nathalie Andersen
- Department of Human Biology, Faculty of Health Sciences, University of Cape Town, Cape Town, 7925, South Africa
- UCT Neuroscience Institute, Cape Town, South Africa
| | - Mubeen Goolam
- Department of Human Biology, Faculty of Health Sciences, University of Cape Town, Cape Town, 7925, South Africa.
- UCT Neuroscience Institute, Cape Town, South Africa.
| |
Collapse
|
6
|
Sibgatullina G, Ramazanova I, Salnikov V, Stepanov A, Voloshina A, Sapunova A, Mustafina A, Petrov K, Samigullin D. Increased endocytosis rate and enhanced lysosomal pathway of silica-coated superparamagnetic nanoparticles into M-HeLa cells compared with cultured primary motor neurons. Histochem Cell Biol 2024; 161:507-519. [PMID: 38597938 DOI: 10.1007/s00418-024-02283-z] [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] [Accepted: 03/21/2024] [Indexed: 04/11/2024]
Abstract
The unique properties of superparamagnetic iron oxide nanoparticles (SPIONs) enable their use as magnetic biosensors, targeted drug delivery, magnetothermia, magnetic resonance imaging, etc. Today, SPIONs are the only type of metal oxide nanoparticles approved for biomedical application. In this work, we analyzed the cellular response to the previously reported luminescent silica coated SPIONs of the two cell types: M-HeLa cells and primary motor neuron culture. Both internalization pathways and intracellular fate of SPIONs have been compared for these cell lines using fluorescence and transmission electron microscopy. We also applied a pharmacological approach to analyze the endocytosis pathways of SPIONs into the investigated cell lines. The penetration of SPIONs into M-HeLa cells is already noticeable within 30 s of incubation through both caveolin-dependent endocytosis and micropinocytosis. However, incubation for a longer time (1 h at least) is required for the internalization of SPIONs into motor neuron culture cells provided by dynamin-dependent endocytosis and macropinocytosis. The intracellular colocalization assay reveals that the lysosomal internalization pathway of SPIONs is also dependent on the cell type. The lysosomal pathway is much more pronounced for M-HeLa cells compared with motor neurons. The emphasized differences in cellular responses of the two cell lines open up new opportunities in the application of SPIONs in the diagnostics and therapy of cancer cells.
Collapse
Affiliation(s)
- Guzel Sibgatullina
- Kazan Institute of Biochemistry and Biophysics, FRC Kazan Scientific Center, Russian Academy of Sciences, P.O. box 261, Kazan, 420111, Russia
| | - Iliza Ramazanova
- Kazan Institute of Biochemistry and Biophysics, FRC Kazan Scientific Center, Russian Academy of Sciences, P.O. box 261, Kazan, 420111, Russia
| | - Vadim Salnikov
- Kazan Institute of Biochemistry and Biophysics, FRC Kazan Scientific Center, Russian Academy of Sciences, P.O. box 261, Kazan, 420111, Russia
| | - Alexey Stepanov
- Arbuzov Institute of Organic and Physical Chemistry, FRC Kazan Scientific Center, Russian Academy of Sciences, Arbuzov Str., 8, 420088, Kazan, Russia
| | - Alexandra Voloshina
- Arbuzov Institute of Organic and Physical Chemistry, FRC Kazan Scientific Center, Russian Academy of Sciences, Arbuzov Str., 8, 420088, Kazan, Russia
| | - Anastasiia Sapunova
- Arbuzov Institute of Organic and Physical Chemistry, FRC Kazan Scientific Center, Russian Academy of Sciences, Arbuzov Str., 8, 420088, Kazan, Russia
| | - Asiya Mustafina
- Arbuzov Institute of Organic and Physical Chemistry, FRC Kazan Scientific Center, Russian Academy of Sciences, Arbuzov Str., 8, 420088, Kazan, Russia
| | - Konstantin Petrov
- Kazan Institute of Biochemistry and Biophysics, FRC Kazan Scientific Center, Russian Academy of Sciences, P.O. box 261, Kazan, 420111, Russia
- Arbuzov Institute of Organic and Physical Chemistry, FRC Kazan Scientific Center, Russian Academy of Sciences, Arbuzov Str., 8, 420088, Kazan, Russia
| | - Dmitry Samigullin
- Kazan Institute of Biochemistry and Biophysics, FRC Kazan Scientific Center, Russian Academy of Sciences, P.O. box 261, Kazan, 420111, Russia.
- Department of Radiophotonics and Microwave Technologies, Kazan National Research Technical University Named After A.N. Tupolev-KAI, 10 K. Marx St., Kazan, 420111, Russia.
| |
Collapse
|
7
|
Thiry L, Sirois J, Durcan TM, Stifani S. Generation of human iPSC-derived phrenic-like motor neurons to model respiratory motor neuron degeneration in ALS. Commun Biol 2024; 7:238. [PMID: 38418587 PMCID: PMC10901792 DOI: 10.1038/s42003-024-05925-z] [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: 07/21/2023] [Accepted: 02/16/2024] [Indexed: 03/01/2024] Open
Abstract
The fatal motor neuron (MN) disease Amyotrophic Lateral Sclerosis (ALS) is characterized by progressive MN degeneration. Phrenic MNs (phMNs) controlling the activity of the diaphragm are prone to degeneration in ALS, leading to death by respiratory failure. Understanding of the mechanisms of phMN degeneration in ALS is limited, mainly because human experimental models to study phMNs are lacking. Here we describe a method enabling the derivation of phrenic-like MNs from human iPSCs (hiPSC-phMNs) within 30 days. This protocol uses an optimized combination of small molecules followed by cell-sorting based on a cell-surface protein enriched in hiPSC-phMNs, and is highly reproducible using several hiPSC lines. We show further that hiPSC-phMNs harbouring ALS-associated amplification of the C9orf72 gene progressively lose their electrophysiological activity and undergo increased death compared to isogenic controls. These studies establish a previously unavailable protocol to generate human phMNs offering a disease-relevant system to study mechanisms of respiratory MN dysfunction.
Collapse
Affiliation(s)
- Louise Thiry
- Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital, McGill University, 3801, rue University, Montreal, QC, H3A 2B4, Canada
- Early Drug Discovery Unit, Montreal Neurological Institute-Hospital, McGill University, 3801, rue University, Montreal, QC, H3A 2B4, Canada
| | - Julien Sirois
- Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital, McGill University, 3801, rue University, Montreal, QC, H3A 2B4, Canada
- Early Drug Discovery Unit, Montreal Neurological Institute-Hospital, McGill University, 3801, rue University, Montreal, QC, H3A 2B4, Canada
| | - Thomas M Durcan
- Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital, McGill University, 3801, rue University, Montreal, QC, H3A 2B4, Canada
- Early Drug Discovery Unit, Montreal Neurological Institute-Hospital, McGill University, 3801, rue University, Montreal, QC, H3A 2B4, Canada
| | - Stefano Stifani
- Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital, McGill University, 3801, rue University, Montreal, QC, H3A 2B4, Canada.
| |
Collapse
|
8
|
Lázaro J, Sochacki J, Ebisuya M. The stem cell zoo for comparative studies of developmental tempo. Curr Opin Genet Dev 2024; 84:102149. [PMID: 38199063 PMCID: PMC10882223 DOI: 10.1016/j.gde.2023.102149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Revised: 12/15/2023] [Accepted: 12/17/2023] [Indexed: 01/12/2024]
Abstract
The rate of development is highly variable across animal species. However, the mechanisms regulating developmental tempo have remained elusive due to difficulties in performing direct interspecies comparisons. Here, we discuss how pluripotent stem cell-based models of development can be used to investigate cell- and tissue-autonomous temporal processes. These systems enable quantitative comparisons of different animal species under similar experimental conditions. Moreover, the constantly growing stem cell zoo collection allows the extension of developmental studies to a great number of unconventional species. We argue that the stem cell zoo constitutes a powerful platform to perform comparative studies of developmental tempo, as well as to study other forms of biological time control such as species-specific lifespan, heart rate, and circadian clocks.
Collapse
Affiliation(s)
- Jorge Lázaro
- European Molecular Biology Laboratory (EMBL) Barcelona, Dr. Aiguader 88, 08003 Barcelona, Spain; Collaboration for joint PhD degree between EMBL and Heidelberg University, Faculty of Biosciences, Heidelberg, Germany. https://twitter.com/@JorgeLazaroF
| | - Jaroslaw Sochacki
- European Molecular Biology Laboratory (EMBL) Barcelona, Dr. Aiguader 88, 08003 Barcelona, Spain
| | - Miki Ebisuya
- European Molecular Biology Laboratory (EMBL) Barcelona, Dr. Aiguader 88, 08003 Barcelona, Spain; Cluster of Excellence Physics of Life, TU Dresden, Arnoldstraße 18, 01307 Dresden, Germany.
| |
Collapse
|
9
|
Kokkorakis N, Douka K, Nalmpanti A, Politis PK, Zagoraiou L, Matsas R, Gaitanou M. Mirk/Dyrk1B controls ventral spinal cord development via Shh pathway. Cell Mol Life Sci 2024; 81:70. [PMID: 38294527 PMCID: PMC10830675 DOI: 10.1007/s00018-023-05097-9] [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: 07/14/2023] [Revised: 12/14/2023] [Accepted: 12/17/2023] [Indexed: 02/01/2024]
Abstract
Cross-talk between Mirk/Dyrk1B kinase and Sonic hedgehog (Shh)/Gli pathway affects physiology and pathology. Here, we reveal a novel role for Dyrk1B in regulating ventral progenitor and neuron subtypes in the embryonic chick spinal cord (SC) via the Shh pathway. Using in ovo gain-and-loss-of-function approaches at E2, we report that Dyrk1B affects the proliferation and differentiation of neuronal progenitors at E4 and impacts on apoptosis specifically in the motor neuron (MN) domain. Especially, Dyrk1B overexpression decreases the numbers of ventral progenitors, MNs, and V2a interneurons, while the pharmacological inhibition of endogenous Dyrk1B kinase activity by AZ191 administration increases the numbers of ventral progenitors and MNs. Mechanistically, Dyrk1B overexpression suppresses Shh, Gli2 and Gli3 mRNA levels, while conversely, Shh, Gli2 and Gli3 transcription is increased in the presence of Dyrk1B inhibitor AZ191 or Smoothened agonist SAG. Most importantly, in phenotype rescue experiments, SAG restores the Dyrk1B-mediated dysregulation of ventral progenitors. Further at E6, Dyrk1B affects selectively the medial lateral motor neuron column (LMCm), consistent with the expression of Shh in this region. Collectively, these observations reveal a novel regulatory function of Dyrk1B kinase in suppressing the Shh/Gli pathway and thus affecting ventral subtypes in the developing spinal cord. These data render Dyrk1B a possible therapeutic target for motor neuron diseases.
Collapse
Affiliation(s)
- N Kokkorakis
- Laboratory of Cellular and Molecular Neurobiology-Stem Cells, Hellenic Pasteur Institute, Athens, Greece
- Division of Animal and Human Physiology, Department of Biology, National and Kapodistrian University of Athens, Athens, Greece
| | - K Douka
- Laboratory of Cellular and Molecular Neurobiology-Stem Cells, Hellenic Pasteur Institute, Athens, Greece
| | - A Nalmpanti
- Laboratory of Cellular and Molecular Neurobiology-Stem Cells, Hellenic Pasteur Institute, Athens, Greece
- Athens International Master's Programme in Neurosciences, Department of Biology, National and Kapodistrian University of Athens, Athens, Greece
| | - P K Politis
- Center of Basic Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
- School of Medicine, European University Cyprus, Nicosia, Cyprus
| | - L Zagoraiou
- School of Medicine, European University Cyprus, Nicosia, Cyprus
| | - R Matsas
- Laboratory of Cellular and Molecular Neurobiology-Stem Cells, Hellenic Pasteur Institute, Athens, Greece
| | - M Gaitanou
- Laboratory of Cellular and Molecular Neurobiology-Stem Cells, Hellenic Pasteur Institute, Athens, Greece.
| |
Collapse
|
10
|
Wang NB, Lende-Dorn BA, Adewumi HO, Beitz AM, Han P, O'Shea TM, Galloway KE. Proliferation history and transcription factor levels drive direct conversion. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.11.26.568736. [PMID: 38077004 PMCID: PMC10705288 DOI: 10.1101/2023.11.26.568736] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/23/2023]
Abstract
The sparse and stochastic nature of reprogramming has obscured our understanding of how transcription factors drive cells to new identities. To overcome this limit, we developed a compact, portable reprogramming system that increases direct conversion of fibroblasts to motor neurons by two orders of magnitude. We show that subpopulations with different reprogramming potentials are distinguishable by proliferation history. By controlling for proliferation history and titrating each transcription factor, we find that conversion correlates with levels of the pioneer transcription factor Ngn2, whereas conversion shows a biphasic response to Lhx3. Increasing the proliferation rate of adult human fibroblasts generates morphologically mature, induced motor neurons at high rates. Using compact, optimized, polycistronic cassettes, we generate motor neurons that graft with the murine central nervous system, demonstrating the potential for in vivo therapies.
Collapse
Affiliation(s)
- Nathan B Wang
- Department of Chemical Engineering, MIT, Cambridge, MA 02139, USA
| | | | - Honour O Adewumi
- Department of Biomedical Engineering, Boston University, Boston, MA 02215, USA
| | - Adam M Beitz
- Department of Chemical Engineering, MIT, Cambridge, MA 02139, USA
| | - Patrick Han
- Department of Chemical Engineering, MIT, Cambridge, MA 02139, USA
| | - Timothy M O'Shea
- Department of Biomedical Engineering, Boston University, Boston, MA 02215, USA
| | - Kate E Galloway
- Department of Chemical Engineering, MIT, Cambridge, MA 02139, USA
| |
Collapse
|
11
|
Nie L, Yao D, Chen S, Wang J, Pan C, Wu D, Liu N, Tang Z. Directional induction of neural stem cells, a new therapy for neurodegenerative diseases and ischemic stroke. Cell Death Discov 2023; 9:215. [PMID: 37393356 DOI: 10.1038/s41420-023-01532-9] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Revised: 06/16/2023] [Accepted: 06/22/2023] [Indexed: 07/03/2023] Open
Abstract
Due to the limited capacity of the adult mammalian brain to self-repair and regenerate, neurological diseases, especially neurodegenerative disorders and stroke, characterized by irreversible cellular damage are often considered as refractory diseases. Neural stem cells (NSCs) play a unique role in the treatment of neurological diseases for their abilities to self-renew and form different neural lineage cells, such as neurons and glial cells. With the increasing understanding of neurodevelopment and advances in stem cell technology, NSCs can be obtained from different sources and directed to differentiate into a specific neural lineage cell phenotype purposefully, making it possible to replace specific cells lost in some neurological diseases, which provides new approaches to treat neurodegenerative diseases as well as stroke. In this review, we outline the advances in generating several neuronal lineage subtypes from different sources of NSCs. We further summarize the therapeutic effects and possible therapeutic mechanisms of these fated specific NSCs in neurological disease models, with special emphasis on Parkinson's disease and ischemic stroke. Finally, from the perspective of clinical translation, we compare the strengths and weaknesses of different sources of NSCs and different methods of directed differentiation, and propose future research directions for directed differentiation of NSCs in regenerative medicine.
Collapse
Affiliation(s)
- Luwei Nie
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China
| | - Dabao Yao
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China
| | - Shiling Chen
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China
| | - Jingyi Wang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China
| | - Chao Pan
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China
| | - Dongcheng Wu
- Department of Biochemistry and Molecular Biology, Wuhan University School of Basic Medical Sciences, Wuhan, 430030, China
- Wuhan Hamilton Biotechnology Co., Ltd., Wuhan, 430030, China
| | - Na Liu
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China.
| | - Zhouping Tang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China.
| |
Collapse
|
12
|
Buchner F, Dokuzluoglu Z, Grass T, Rodriguez-Muela N. Spinal Cord Organoids to Study Motor Neuron Development and Disease. Life (Basel) 2023; 13:1254. [PMID: 37374039 PMCID: PMC10303776 DOI: 10.3390/life13061254] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Accepted: 05/18/2023] [Indexed: 06/29/2023] Open
Abstract
Motor neuron diseases (MNDs) are a heterogeneous group of disorders that affect the cranial and/or spinal motor neurons (spMNs), spinal sensory neurons and the muscular system. Although they have been investigated for decades, we still lack a comprehensive understanding of the underlying molecular mechanisms; and therefore, efficacious therapies are scarce. Model organisms and relatively simple two-dimensional cell culture systems have been instrumental in our current knowledge of neuromuscular disease pathology; however, in the recent years, human 3D in vitro models have transformed the disease-modeling landscape. While cerebral organoids have been pursued the most, interest in spinal cord organoids (SCOs) is now also increasing. Pluripotent stem cell (PSC)-based protocols to generate SpC-like structures, sometimes including the adjacent mesoderm and derived skeletal muscle, are constantly being refined and applied to study early human neuromuscular development and disease. In this review, we outline the evolution of human PSC-derived models for generating spMN and recapitulating SpC development. We also discuss how these models have been applied to exploring the basis of human neurodevelopmental and neurodegenerative diseases. Finally, we provide an overview of the main challenges to overcome in order to generate more physiologically relevant human SpC models and propose some exciting new perspectives.
Collapse
Affiliation(s)
- Felix Buchner
- German Center for Neurodegenerative Diseases, 01307 Dresden, Germany; (F.B.); (Z.D.); (T.G.)
| | - Zeynep Dokuzluoglu
- German Center for Neurodegenerative Diseases, 01307 Dresden, Germany; (F.B.); (Z.D.); (T.G.)
| | - Tobias Grass
- German Center for Neurodegenerative Diseases, 01307 Dresden, Germany; (F.B.); (Z.D.); (T.G.)
| | - Natalia Rodriguez-Muela
- German Center for Neurodegenerative Diseases, 01307 Dresden, Germany; (F.B.); (Z.D.); (T.G.)
- Center for Regenerative Therapies Dresden, Technische Universität Dresden, 01307 Dresden, Germany
- Max Planck Institute for Molecular Cell Biology and Genetics, 01307 Dresden, Germany
| |
Collapse
|
13
|
Cheung DL, Toda T, Narushima M, Eto K, Takayama C, Ooba T, Wake H, Moorhouse AJ, Nabekura J. KCC2 downregulation after sciatic nerve injury enhances motor function recovery. Sci Rep 2023; 13:7871. [PMID: 37188694 DOI: 10.1038/s41598-023-34701-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2022] [Accepted: 05/05/2023] [Indexed: 05/17/2023] Open
Abstract
Injury to mature neurons induces downregulated KCC2 expression and activity, resulting in elevated intracellular [Cl-] and depolarized GABAergic signaling. This phenotype mirrors immature neurons wherein GABA-evoked depolarizations facilitate neuronal circuit maturation. Thus, injury-induced KCC2 downregulation is broadly speculated to similarly facilitate neuronal circuit repair. We test this hypothesis in spinal cord motoneurons injured by sciatic nerve crush, using transgenic (CaMKII-KCC2) mice wherein conditional CaMKIIα promoter-KCC2 expression coupling selectively prevents injury-induced KCC2 downregulation. We demonstrate, via an accelerating rotarod assay, impaired motor function recovery in CaMKII-KCC2 mice relative to wild-type mice. Across both cohorts, we observe similar motoneuron survival and re-innervation rates, but differing post-injury reorganization patterns of synaptic input to motoneuron somas-for wild-type, both VGLUT1-positive (excitatory) and GAD67-positive (inhibitory) terminal counts decrease; for CaMKII-KCC2, only VGLUT1-positive terminal counts decrease. Finally, we recapitulate the impaired motor function recovery of CaMKII-KCC2 mice in wild-type mice by administering local spinal cord injections of bicuculline (GABAA receptor blockade) or bumetanide (lowers intracellular [Cl-] by NKCC1 blockade) during the early post-injury period. Thus, our results provide direct evidence that injury-induced KCC2 downregulation enhances motor function recovery and suggest an underlying mechanism of depolarizing GABAergic signaling driving adaptive reconfiguration of presynaptic GABAergic input.
Collapse
Affiliation(s)
- Dennis Lawrence Cheung
- Division of Homeostatic Development, National Institute for Physiological Sciences, Okazaki, Aichi, Japan
| | - Takuya Toda
- Division of Homeostatic Development, National Institute for Physiological Sciences, Okazaki, Aichi, Japan
| | - Madoka Narushima
- Division of Homeostatic Development, National Institute for Physiological Sciences, Okazaki, Aichi, Japan
| | - Kei Eto
- Division of Homeostatic Development, National Institute for Physiological Sciences, Okazaki, Aichi, Japan
- Department of Physiology, School of Allied Health Sciences, Kitasato University, Sagamihara, Kanagawa, Japan
| | | | - Tatsuko Ooba
- Division of Homeostatic Development, National Institute for Physiological Sciences, Okazaki, Aichi, Japan
| | - Hiroaki Wake
- Division of Multicellular Circuit Dynamics, National Institute for Physiological Sciences, Okazaki, Aichi, Japan
- Graduate School of Medicine, Nagoya University, Nagoya, Aichi, Japan
| | - Andrew John Moorhouse
- School of Biomedical Sciences, UNSW Sydney (The University of New South Wales), Sydney, New South Wales, Australia
| | - Junichi Nabekura
- Division of Homeostatic Development, National Institute for Physiological Sciences, Okazaki, Aichi, Japan.
- Graduate School of Medicine, Nagoya University, Nagoya, Aichi, Japan.
- School of Life Science, SOKENDAI (The Graduate University for Advanced Studies), Okazaki, Aichi, Japan.
| |
Collapse
|
14
|
Xu HJ, Yao Y, Yao F, Chen J, Li M, Yang X, Li S, Lu F, Hu P, He S, Peng G, Jing N. Generation of functional posterior spinal motor neurons from hPSCs-derived human spinal cord neural progenitor cells. CELL REGENERATION (LONDON, ENGLAND) 2023; 12:15. [PMID: 36949352 PMCID: PMC10033800 DOI: 10.1186/s13619-023-00159-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Accepted: 02/24/2023] [Indexed: 03/24/2023]
Abstract
Spinal motor neurons deficiency results in a series of devastating disorders such as amyotrophic lateral sclerosis (ALS), spinal muscular atrophy (SMA) and spinal cord injury (SCI). These disorders are currently incurable, while human pluripotent stem cells (hPSCs)-derived spinal motor neurons are promising but suffered from inappropriate regional identity and functional immaturity for the study and treatment of posterior spinal cord related injuries. In this study, we have established human spinal cord neural progenitor cells (hSCNPCs) via hPSCs differentiated neuromesodermal progenitors (NMPs) and demonstrated the hSCNPCs can be continuously expanded up to 40 passages. hSCNPCs can be rapidly differentiated into posterior spinal motor neurons with high efficiency. The functional maturity has been examined in detail. Moreover, a co-culture scheme which is compatible for both neural and muscular differentiation is developed to mimic the neuromuscular junction (NMJ) formation in vitro. Together, these studies highlight the potential avenues for generating clinically relevant spinal motor neurons and modeling neuromuscular diseases through our defined hSCNPCs.
Collapse
Affiliation(s)
- He Jax Xu
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai, 200031, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Yao Yao
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai, 200031, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Fenyong Yao
- School of Life Science and Technology, ShanghaiTech University, Shanghai, 201210, China
| | - Jiehui Chen
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai, 200031, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Meishi Li
- University of Chinese Academy of Sciences, Beijing, China
- Center for Cell Lineage and Development, CAS Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, GIBH-HKU Guangdong-Hong Kong Stem Cell and Regenerative Medicine Research Centre, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China
| | - Xianfa Yang
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai, 200031, China
- University of Chinese Academy of Sciences, Beijing, China
- Guangzhou Laboratory/Bioland Laboratory, Guangzhou, 510005, China
| | - Sheng Li
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai, 200031, China
- Xinhua Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 20023, China
| | - Fangru Lu
- Center for Cell Lineage and Development, CAS Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, GIBH-HKU Guangdong-Hong Kong Stem Cell and Regenerative Medicine Research Centre, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China
| | - Ping Hu
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai, 200031, China
- University of Chinese Academy of Sciences, Beijing, China
- Guangzhou Laboratory/Bioland Laboratory, Guangzhou, 510005, China
- Xinhua Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 20023, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, 100101, China
| | - Shuijin He
- School of Life Science and Technology, ShanghaiTech University, Shanghai, 201210, China.
| | - Guangdun Peng
- University of Chinese Academy of Sciences, Beijing, China.
- Center for Cell Lineage and Development, CAS Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, GIBH-HKU Guangdong-Hong Kong Stem Cell and Regenerative Medicine Research Centre, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China.
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, 100101, China.
- Center for Cell Lineage and Atlas, Bioland Laboratory, Guangzhou, 510005, China.
| | - Naihe Jing
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai, 200031, China.
- University of Chinese Academy of Sciences, Beijing, China.
- Guangzhou Laboratory/Bioland Laboratory, Guangzhou, 510005, China.
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, 100101, China.
| |
Collapse
|
15
|
Limone F, Guerra San Juan I, Mitchell JM, Smith JLM, Raghunathan K, Meyer D, Ghosh SD, Couto A, Klim JR, Joseph BJ, Gold J, Mello CJ, Nemesh J, Smith BM, Verhage M, McCarroll SA, Pietiläinen O, Nehme R, Eggan K. Efficient generation of lower induced motor neurons by coupling Ngn2 expression with developmental cues. Cell Rep 2023; 42:111896. [PMID: 36596304 PMCID: PMC10117176 DOI: 10.1016/j.celrep.2022.111896] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Revised: 06/01/2022] [Accepted: 12/08/2022] [Indexed: 01/03/2023] Open
Abstract
Human pluripotent stem cells (hPSCs) are a powerful tool for disease modeling of hard-to-access tissues (such as the brain). Current protocols either direct neuronal differentiation with small molecules or use transcription-factor-mediated programming. In this study, we couple overexpression of transcription factor Neurogenin2 (Ngn2) with small molecule patterning to differentiate hPSCs into lower induced motor neurons (liMoNes/liMNs). This approach induces canonical MN markers including MN-specific Hb9/MNX1 in more than 95% of cells. liMNs resemble bona fide hPSC-derived MN, exhibit spontaneous electrical activity, express synaptic markers, and can contact muscle cells in vitro. Pooled, multiplexed single-cell RNA sequencing on 50 hPSC lines reveals reproducible populations of distinct subtypes of cervical and brachial MNs that resemble their in vivo, embryonic counterparts. Combining small molecule patterning with Ngn2 overexpression facilitates high-yield, reproducible production of disease-relevant MN subtypes, which is fundamental in propelling our knowledge of MN biology and its disruption in disease.
Collapse
Affiliation(s)
- Francesco Limone
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138, USA; Stanley Centre for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Leiden University Medical Center, LUMC, 2333 ZA Leiden, the Netherlands.
| | - Irune Guerra San Juan
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138, USA; Stanley Centre for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Department of Functional Genomics, Center for Neurogenomics and Cognitive Research, Vrije Universiteit, Amsterdam, the Netherlands; Human Genetics, Amsterdam University Medical Center, Amsterdam, the Netherlands
| | - Jana M Mitchell
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138, USA; Stanley Centre for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Janell L M Smith
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138, USA
| | - Kavya Raghunathan
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138, USA; Stanley Centre for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Daniel Meyer
- Stanley Centre for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
| | - Sulagna Dia Ghosh
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138, USA; Stanley Centre for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
| | - Alexander Couto
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138, USA
| | - Joseph R Klim
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138, USA; Stanley Centre for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Brian J Joseph
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138, USA; Departments of Pathology and Cell Biology, Columbia University Irving Medical Centre, New York, NY 10032, USA
| | - John Gold
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138, USA
| | - Curtis J Mello
- Stanley Centre for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
| | - James Nemesh
- Stanley Centre for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
| | - Brittany M Smith
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138, USA
| | - Matthijs Verhage
- Department of Functional Genomics, Center for Neurogenomics and Cognitive Research, Vrije Universiteit, Amsterdam, the Netherlands; Human Genetics, Amsterdam University Medical Center, Amsterdam, the Netherlands
| | - Steven A McCarroll
- Stanley Centre for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
| | - Olli Pietiläinen
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138, USA; Stanley Centre for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Neuroscience Center, Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland
| | - Ralda Nehme
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138, USA; Stanley Centre for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Kevin Eggan
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138, USA.
| |
Collapse
|
16
|
Castillo Bautista CM, Sterneckert J. Progress and challenges in directing the differentiation of human iPSCs into spinal motor neurons. Front Cell Dev Biol 2023; 10:1089970. [PMID: 36684437 PMCID: PMC9849822 DOI: 10.3389/fcell.2022.1089970] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Accepted: 12/21/2022] [Indexed: 01/07/2023] Open
Abstract
Motor neuron (MN) diseases, including amyotrophic lateral sclerosis, progressive bulbar palsy, primary lateral sclerosis and spinal muscular atrophy, cause progressive paralysis and, in many cases, death. A better understanding of the molecular mechanisms of pathogenesis is urgently needed to identify more effective therapies. However, studying MNs has been extremely difficult because they are inaccessible in the spinal cord. Induced pluripotent stem cells (iPSCs) can generate a theoretically limitless number of MNs from a specific patient, making them powerful tools for studying MN diseases. However, to reach their potential, iPSCs need to be directed to efficiently differentiate into functional MNs. Here, we review the reported differentiation protocols for spinal MNs, including induction with small molecules, expression of lineage-specific transcription factors, 2-dimensional and 3-dimensional cultures, as well as the implementation of microfluidics devices and co-cultures with other cell types, including skeletal muscle. We will summarize the advantages and disadvantages of each strategy. In addition, we will provide insights into how to address some of the remaining challenges, including reproducibly obtaining mature and aged MNs.
Collapse
Affiliation(s)
| | - Jared Sterneckert
- Center for Regenerative Therapies TU Dresden (CRTD), Technische Universität (TU) Dresden, Dresden, Germany,Medical Faculty Carl Gustav Carus of TU Dresden, Dresden, Germany,*Correspondence: Jared Sterneckert,
| |
Collapse
|
17
|
Wu J, Tang Y. Transcription Factor-Mediated Differentiation of Motor Neurons from Human Pluripotent Stem Cells. Methods Mol Biol 2023; 2593:245-258. [PMID: 36513936 DOI: 10.1007/978-1-0716-2811-9_16] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
Studying the pathogenesis of neurological diseases with animal models might not always truly recapitulate their pathophysiology, due to species differences. Fortunately, human pluripotent stem cells (hPSCs) including embryonic stem cells (ESCs) and induced pluripotent stem cells (iPSCs), especially derived from patients, have been widely employed to induce neural progenitor cells (NPCs) and further multiple neural subtypes. Particularly in the past decade, hPSC-based cell sources have been applied in studying neural development, cell therapy, disease modeling, and drug screening, among others. The generation of unlimited amount of neurons also facilitates a variety of biochemical assays, mass spectrometry, omic analysis, and next-generation sequencing, which thus provides an excellent tool in modeling neurodegenerative and neurodevelopmental diseases. Dysfunction or death of motor neurons (MNs) in the spinal cord and motor cortex is implicated in various motor neuron diseases (MNDs). Yet, producing high-purity and high-yield MNs remains a major challenge due to the complexity of MN specification during development. In this chapter, we describe a method of generating functional MNs via lentiviral delivery of transcription factors, based on the preservable NPC platform derived from hPSCs. Specifically, we transduce NPCs with a single lentivirus co-expressing three transcription factors including NGN2, ISL1, and LHX3, which is necessary and sufficient to induce mature MNs with high efficiencies (~90%) within 3 weeks. This chapter thus provides a robust method to generate high-purity hPSC-MNs at very high yields, enabling the acquisition of rich patient-specific MNs to be used for modeling the molecular underpinnings of MNDs.
Collapse
Affiliation(s)
- Junjiao Wu
- Department of Rheumatology and Immunology, Xiangya Hospital, Central South University, Changsha, China
- Provincial Clinical Research Center for Rheumatic and Immunologic Diseases, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Yu Tang
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China.
- Department of Geriatrics, Xiangya Hospital, Central South University, Changsha, China.
- Aging Research Center, Xiangya Hospital, Central South University, Changsha, China.
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan, China.
- The Biobank of Xiangya Hospital, Central South University, Changsha, Hunan, China.
| |
Collapse
|
18
|
Gao Y. Using Human iPSC-Derived Peripheral Nervous System Disease Models for Drug Discovery. Handb Exp Pharmacol 2023; 281:191-205. [PMID: 37815594 DOI: 10.1007/164_2023_690] [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] [Indexed: 10/11/2023]
Abstract
Induced pluripotent stem cells (IPSCs), with their remarkable ability to differentiate into various cell types, including peripheral nervous system cells such as neurons and glial cells, offer an excellent platform for in vitro disease modeling. These iPSC-derived disease models have proven valuable in drug discovery, as they provide more precise simulations of a patient's disease state and allow for the assessment of potential therapeutic effectiveness and safety.
Collapse
Affiliation(s)
- Yuan Gao
- Translational Medicine Research Center, Shanxi Medical University, Taiyuan, China
| |
Collapse
|
19
|
Schaefers C, Rothmiller S, Thiermann H, Rein T, Schmidt A. The Efficiency of Direct Maturation: the Comparison of Two hiPSC Differentiation Approaches into Motor Neurons. Stem Cells Int 2022; 2022:1320950. [PMID: 36530489 PMCID: PMC9757946 DOI: 10.1155/2022/1320950] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2022] [Revised: 11/10/2022] [Accepted: 11/11/2022] [Indexed: 02/23/2025] Open
Abstract
Motor neurons (MNs) derived from human-induced pluripotent stem cells (hiPSC) hold great potential for the treatment of various motor neurodegenerative diseases as transplantations with a low-risk of rejection are made possible. There are many hiPSC differentiation protocols that pursue to imitate the multistep process of motor neurogenesis in vivo. However, these often apply viral vectors, feeder cells, or antibiotics to generate hiPSC and MNs, limiting their translational potential. In this study, a virus-, feeder-, and antibiotic-free method was used for reprogramming hiPSC, which were maintained in culture medium produced under clinical good manufacturing practice. Differentiation into MNs was performed with standardized, chemically defined, and antibiotic-free culture media. The identity of hiPSC, neuronal progenitors, and mature MNs was continuously verified by the detection of specific markers at the genetic and protein level via qRT-PCR, flow cytometry, Western Blot, and immunofluorescence. MNX1- and ChAT-positive motoneuronal progenitor cells were formed after neural induction via dual-SMAD inhibition and expansion. For maturation, an approach aiming to directly mature these progenitors was compared to an approach that included an additional differentiation step for further specification. Although both approaches generated mature MNs expressing characteristic postmitotic markers, the direct maturation approach appeared to be more efficient. These results provide new insights into the suitability of two standardized differentiation approaches for generating mature MNs, which might pave the way for future clinical applications.
Collapse
Affiliation(s)
- Catherine Schaefers
- Bundeswehr Institute of Pharmacology and Toxicology, Neuherbergstr. 11, 80937 Munich, Germany
| | - Simone Rothmiller
- Bundeswehr Institute of Pharmacology and Toxicology, Neuherbergstr. 11, 80937 Munich, Germany
| | - Horst Thiermann
- Bundeswehr Institute of Pharmacology and Toxicology, Neuherbergstr. 11, 80937 Munich, Germany
| | - Theo Rein
- Max Planck Institute of Psychiatry, Kraepelinstr. 2-10, 80804 Munich, Germany
| | - Annette Schmidt
- Bundeswehr Institute of Pharmacology and Toxicology, Neuherbergstr. 11, 80937 Munich, Germany
- Institute of Sport Science, University of the Bundeswehr Munich, Werner-Heisenberg-Weg 39, 85577 Neubiberg, Germany
| |
Collapse
|
20
|
Limone F, Klim JR, Mordes DA. Pluripotent stem cell strategies for rebuilding the human brain. Front Aging Neurosci 2022; 14:1017299. [PMID: 36408113 PMCID: PMC9667068 DOI: 10.3389/fnagi.2022.1017299] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Accepted: 09/27/2022] [Indexed: 01/03/2023] Open
Abstract
Neurodegenerative disorders have been extremely challenging to treat with traditional drug-based approaches and curative therapies are lacking. Given continued progress in stem cell technologies, cell replacement strategies have emerged as concrete and potentially viable therapeutic options. In this review, we cover advances in methods used to differentiate human pluripotent stem cells into several highly specialized types of neurons, including cholinergic, dopaminergic, and motor neurons, and the potential clinical applications of stem cell-derived neurons for common neurodegenerative diseases, including Alzheimer's disease, Parkinson's disease, Huntington's disease, ataxia, and amyotrophic lateral sclerosis. Additionally, we summarize cellular differentiation techniques for generating glial cell populations, including oligodendrocytes and microglia, and their conceivable translational roles in supporting neural function. Clinical trials of specific cell replacement therapies in the nervous system are already underway, and several attractive avenues in regenerative medicine warrant further investigation.
Collapse
Affiliation(s)
- Francesco Limone
- Department of Stem Cell and Regenerative Biology, Harvard Stem Cell Institute, Cambridge, MA, United States
- Department of Molecular and Cellular Biology, Harvard Stem Cell Institute, Cambridge, MA, United States
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, United States
- Leiden University Medical Center, Leiden, Netherlands
| | | | - Daniel A. Mordes
- Institute for Neurodegenerative Diseases, Department of Pathology, University of California, San Francisco, San Francisco, CA, United States
| |
Collapse
|
21
|
Nango H, Kosuge Y. Present State and Future Perspectives of Prostaglandins as a Differentiation Factor in Motor Neurons. Cell Mol Neurobiol 2022; 42:2097-2108. [PMID: 34032949 PMCID: PMC11421640 DOI: 10.1007/s10571-021-01104-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Accepted: 05/18/2021] [Indexed: 11/28/2022]
Abstract
Spinal motor neurons have the longest axons that innervate the skeletal muscles of the central nervous system. Motor neuron diseases caused by spinal motor neuron cell death are incurable due to the unique and irreplaceable nature of their neural circuits. Understanding the mechanisms of neurogenesis, neuritogenesis, and synaptogenesis in motor neurons will allow investigators to develop new in vitro models and regenerative therapies for motor neuron diseases. In particular, small molecules can directly reprogram and convert into neural stem cells and neurons, and promote neuron-like cell differentiation. Prostaglandins are known to have a role in the differentiation and tissue regeneration of several cell types and organs. However, the involvement of prostaglandins in the differentiation of motor neurons from neural stem cells is poorly understood. The general cell line used in research on motor neuron diseases is the mouse neuroblastoma and spinal motor neuron fusion cell line NSC-34. Recently, our laboratory reported that prostaglandin E2 and prostaglandin D2 enhanced the conversion of NSC-34 cells into motor neuron-like cells with neurite outgrowth. Moreover, we found that prostaglandin E2-differentiated NSC-34 cells had physiological and electrophysiological properties of mature motor neurons. In this review article, we provide contemporary evidence on the effects of prostaglandins, particularly prostaglandin E2 and prostaglandin D2, on differentiation and neural conversion. We also discuss the potential of prostaglandins as candidates for the development of new therapeutic drugs for motor neuron diseases.
Collapse
Affiliation(s)
- Hiroshi Nango
- Laboratory of Pharmacology, School of Pharmacy, Nihon University, 7-7-1 Narashinodai, Funabashi-shi, Chiba, 274-8555, Japan
| | - Yasuhiro Kosuge
- Laboratory of Pharmacology, School of Pharmacy, Nihon University, 7-7-1 Narashinodai, Funabashi-shi, Chiba, 274-8555, Japan.
| |
Collapse
|
22
|
Rosholm KR, Badone B, Karatsiompani S, Nagy D, Seibertz F, Voigt N, Bell DC. Adventures and Advances in Time Travel With Induced Pluripotent Stem Cells and Automated Patch Clamp. Front Mol Neurosci 2022; 15:898717. [PMID: 35813069 PMCID: PMC9258620 DOI: 10.3389/fnmol.2022.898717] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Accepted: 05/13/2022] [Indexed: 01/21/2023] Open
Abstract
In the Hollywood blockbuster “The Curious Case of Benjamin Button” a fantastical fable unfolds of a man’s life that travels through time reversing the aging process; as the tale progresses, the frail old man becomes a vigorous, vivacious young man, then man becomes boy and boy becomes baby. The reality of cellular time travel, however, is far more wondrous: we now have the ability to both reverse and then forward time on mature cells. Four proteins were found to rewind the molecular clock of adult cells back to their embryonic, “blank canvas” pluripotent stem cell state, allowing these pluripotent stem cells to then be differentiated to fast forward their molecular clocks to the desired adult specialist cell types. These four proteins – the “Yamanaka factors” – form critical elements of this cellular time travel, which deservedly won Shinya Yamanaka the Nobel Prize for his lab’s work discovering them. Human induced pluripotent stem cells (hiPSCs) hold much promise in our understanding of physiology and medicine. They encapsulate the signaling pathways of the desired cell types, such as cardiomyocytes or neurons, and thus act as model cells for defining the critical ion channel activity in healthy and disease states. Since hiPSCs can be derived from any patient, highly specific, personalized (or stratified) physiology, and/or pathophysiology can be defined, leading to exciting developments in personalized medicines and interventions. As such, hiPSC married with high throughput automated patch clamp (APC) ion channel recording platforms provide a foundation for significant physiological, medical and drug discovery advances. This review aims to summarize the current state of affairs of hiPSC and APC: the background and recent advances made; and the pros, cons and challenges of these technologies. Whilst the authors have yet to finalize a fully functional time traveling machine, they will endeavor to provide plausible future projections on where hiPSC and APC are likely to carry us. One future projection the authors are confident in making is the increasing necessity and adoption of these technologies in the discovery of the next blockbuster, this time a life-enhancing ion channel drug, not a fantastical movie.
Collapse
Affiliation(s)
- Kadla R. Rosholm
- Sophion Bioscience A/S, Ballerup, Denmark
- *Correspondence: Kadla R. Rosholm,
| | | | | | - David Nagy
- Sophion Bioscience Inc., Woburn, MA, United States
| | - Fitzwilliam Seibertz
- Institute of Pharmacology and Toxicology, University Medical Center Göttingen, Göttingen, Germany
- German Center for Cardiovascular Research (DZHK), Göttingen, Germany
| | - Niels Voigt
- Institute of Pharmacology and Toxicology, University Medical Center Göttingen, Göttingen, Germany
- German Center for Cardiovascular Research (DZHK), Göttingen, Germany
- Cluster of Excellence “Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells” (MBExC), University of Göttingen, Göttingen, Germany
| | | |
Collapse
|
23
|
Ding Q, Kesavan K, Lee KM, Wimberger E, Robertson T, Gill M, Power D, Chang J, Fard AT, Mar JC, Henderson RD, Heggie S, McCombe PA, Jeffree RL, Colditz MJ, Hilliard MA, Ng DCH, Steyn FJ, Phillips WD, Wolvetang EJ, Ngo ST, Noakes PG. Impaired signaling for neuromuscular synaptic maintenance is a feature of Motor Neuron Disease. Acta Neuropathol Commun 2022; 10:61. [PMID: 35468848 PMCID: PMC9040261 DOI: 10.1186/s40478-022-01360-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Accepted: 04/06/2022] [Indexed: 11/17/2022] Open
Abstract
A central event in the pathogenesis of motor neuron disease (MND) is the loss of neuromuscular junctions (NMJs), yet the mechanisms that lead to this event in MND remain to be fully elucidated. Maintenance of the NMJ relies upon neural agrin (n-agrin) which, when released from the nerve terminal, activates the postsynaptic Muscle Specific Kinase (MuSK) signaling complex to stabilize clusters of acetylcholine receptors. Here, we report that muscle from MND patients has an increased proportion of slow fibers and muscle fibers with smaller diameter. Muscle cells cultured from MND biopsies failed to form large clusters of acetylcholine receptors in response to either non-MND human motor axons or n-agrin. Furthermore, levels of expression of MuSK, and MuSK-complex components: LRP4, Caveolin-3, and Dok7 differed between muscle cells cultured from MND patients compared to those from non-MND controls. To our knowledge, this is the first time a fault in the n-agrin-LRP4-MuSK signaling pathway has been identified in muscle from MND patients. Our results highlight the n-agrin-LRP4-MuSK signaling pathway as a potential therapeutic target to prolong muscle function in MND.
Collapse
|
24
|
Bazarek S, Johnston BR, Sten M, Mandeville R, Eggan K, Wainger BJ, Brown JM. Spinal motor neuron transplantation to enhance nerve reconstruction strategies: Towards a cell therapy. Exp Neurol 2022; 353:114054. [PMID: 35341748 DOI: 10.1016/j.expneurol.2022.114054] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2022] [Revised: 03/16/2022] [Accepted: 03/21/2022] [Indexed: 11/19/2022]
Abstract
Nerve transfers have become a powerful intervention to restore function following devastating paralyzing injuries. A major limitation to peripheral nerve repair and reconstructive strategies is the progressive, fibrotic degeneration of the distal nerve and denervated muscle, eventually precluding recovery of these targets and thus defining a time window within which reinnervation must occur. One proven strategy in the clinic has been the sacrifice and transfer of an adjacent distal motor nerve to provide axons to occupy, and thus preserve (or "babysit"), the target muscle. However, available nearby nerves are limited in severe brachial plexus or spinal cord injury. An alternative and novel proposition is the transplantation of spinal motor neurons (SMNs) derived from human induced pluripotent stem cells (iPSCs) into the target nerve to extend their axons to occupy and preserve the targets. These cells could potentially be delivered through minimally invasive or percutaneous techniques. Several reports have demonstrated survival, functional innervation, and muscular preservation following transplantation of SMNs into rodent nerves. Advances in the generation, culture, and differentiation of human iPSCs now offer the possibility for an unlimited supply of clinical grade SMNs. This review will discuss the previous reports of peripheral SMN transplantation, outline key considerations, and propose next steps towards advancing this approach to clinic. Stem cells have garnered great enthusiasm for their potential to revolutionize medicine. However, this excitement has often led to premature clinical studies with ill-defined cell products and mechanisms of action, particularly in spinal cord injury. We believe the peripheral transplantation of a defined SMN population to address neuromuscular degeneration will be transformative in augmenting current reconstructive strategies. By thus removing the current barriers of time and distance, this strategy would dramatically enhance the potential for reconstruction and functional recovery in otherwise hopeless paralyzing injuries. Furthermore, this strategy may be used as a permanent axon replacement following destruction of lower motor neurons and would enable exogenous stimulation options, such as pacing of transplanted SMN axons in the phrenic nerve to avoid mechanical ventilation in high cervical cord injury or amyotrophic lateral sclerosis.
Collapse
Affiliation(s)
- Stanley Bazarek
- Department of Neurosurgery, Brigham & Women's Hospital, Harvard Medical School, Boston, MA, United States of America; Department of Neurosurgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States of America
| | - Benjamin R Johnston
- Department of Neurosurgery, Brigham & Women's Hospital, Harvard Medical School, Boston, MA, United States of America; Department of Neurosurgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States of America
| | - Margaret Sten
- Department of Neurosurgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States of America
| | - Ross Mandeville
- Department of Neurology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States of America
| | - Kevin Eggan
- BioMarin Pharmaceutical Inc., San Rafael, CA, United States of America
| | - Brian J Wainger
- Departments of Neurology and Anesthesia, Critical Care & Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States of America.
| | - Justin M Brown
- Department of Neurosurgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States of America.
| |
Collapse
|
25
|
Alvarez-Dominguez JR, Melton DA. Cell maturation: Hallmarks, triggers, and manipulation. Cell 2022; 185:235-249. [PMID: 34995481 PMCID: PMC8792364 DOI: 10.1016/j.cell.2021.12.012] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Revised: 12/03/2021] [Accepted: 12/10/2021] [Indexed: 02/06/2023]
Abstract
How cells become specialized, or "mature," is important for cell and developmental biology. While maturity is usually deemed a terminal fate, it may be more helpful to consider maturation not as a switch but as a dynamic continuum of adaptive phenotypic states set by genetic and environment programing. The hallmarks of maturity comprise changes in anatomy (form, gene circuitry, and interconnectivity) and physiology (function, rhythms, and proliferation) that confer adaptive behavior. We discuss efforts to harness their chemical (nutrients, oxygen, and growth factors) and physical (mechanical, spatial, and electrical) triggers in vitro and in vivo and how maturation strategies may support disease research and regenerative medicine.
Collapse
Affiliation(s)
- Juan R. Alvarez-Dominguez
- Department of Cell and Developmental Biology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Douglas A. Melton
- Department of Stem Cell and Regenerative Biology, Harvard Stem Cell Institute, Harvard University, Cambridge, MA 02138, USA
| |
Collapse
|
26
|
Will Cannabigerol Trigger Neuroregeneration after a Spinal Cord Injury? An In Vitro Answer from NSC-34 Scratch-Injured Cells Transcriptome. Pharmaceuticals (Basel) 2022; 15:ph15020117. [PMID: 35215230 PMCID: PMC8875351 DOI: 10.3390/ph15020117] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 01/12/2022] [Accepted: 01/17/2022] [Indexed: 02/07/2023] Open
Abstract
Spinal cord injury affects the lives of millions of people around the world, often causing disability and, in unfortunate circumstances, death. Rehabilitation can partly improve outcomes and only a small percentage of patients, typically the least injured, can hope to return to normal living conditions. Cannabis sativa is gaining more and more interest in recent years, even though its beneficial properties have been known for thousands of years. Cannabigerol (CBG), extracted from C. sativa, is defined as the “mother of all cannabinoids” and its properties range from anti-inflammatory to antioxidant and neuroprotection. Using NSC-34 cells to model spinal cord injury in vitro, our work evaluated the properties of CBG treatments in motor neuron regeneration. While pre-treatment can modulate oxidative stress and increase antioxidant enzyme genes, such as Tnx1, decreasing Nos1 post-treatment seems to induce regeneration genes by triggering different pathways, such as Gap43 via p53 acetylation by Ep300 and Ddit3 and Xbp1 via Bdnf signaling, along with cytoskeletal remodeling signaling genes Nrp1 and Map1b. Our results indicate CBG as a phytocompound worth further investigation in the field of neuronal regeneration.
Collapse
|
27
|
Bos R, Rihan K, Quintana P, El-Bazzal L, Bernard-Marissal N, Da Silva N, Jabbour R, Mégarbané A, Bartoli M, Brocard F, Delague V. Altered action potential waveform and shorter axonal initial segment in hiPSC-derived motor neurons with mutations in VRK1. Neurobiol Dis 2022; 164:105609. [PMID: 34990802 DOI: 10.1016/j.nbd.2021.105609] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Revised: 10/24/2021] [Accepted: 12/30/2021] [Indexed: 11/25/2022] Open
Abstract
We recently described new pathogenic variants in VRK1, in patients affected with distal Hereditary Motor Neuropathy associated with upper motor neurons signs. Specifically, we provided evidences that hiPSC-derived Motor Neurons (hiPSC-MN) from these patients display Cajal Bodies (CBs) disassembly and defects in neurite outgrowth and branching. We here focused on the Axonal Initial Segment (AIS) and the related firing properties of hiPSC-MNs from these patients. We found that the patient's Action Potential (AP) was smaller in amplitude, larger in duration, and displayed a more depolarized threshold while the firing patterns were not altered. These alterations were accompanied by a decrease in the AIS length measured in patients' hiPSC-MNs. These data indicate that mutations in VRK1 impact the AP waveform and the AIS organization in MNs and may ultimately lead to the related motor neuron disease.
Collapse
Affiliation(s)
- Rémi Bos
- Aix Marseille Univ, CNRS, Institut de Neurosciences de la Timone (INT), UMR 7289, Marseille, France.
| | - Khalil Rihan
- Aix Marseille Univ, Inserm, MMG, U 1251, Institut Marseille Maladies Rares (MarMaRa), Marseille, France
| | - Patrice Quintana
- Aix Marseille Univ, Inserm, MMG, U 1251, Institut Marseille Maladies Rares (MarMaRa), Marseille, France
| | - Lara El-Bazzal
- Aix Marseille Univ, Inserm, MMG, U 1251, Institut Marseille Maladies Rares (MarMaRa), Marseille, France
| | - Nathalie Bernard-Marissal
- Aix Marseille Univ, Inserm, MMG, U 1251, Institut Marseille Maladies Rares (MarMaRa), Marseille, France
| | - Nathalie Da Silva
- Aix Marseille Univ, Inserm, MMG, U 1251, Institut Marseille Maladies Rares (MarMaRa), Marseille, France
| | - Rosette Jabbour
- Neurology Division, Department of Internal Medicine, St George Hospital University Medical Center, University of Balamand, Beirut, Lebanon
| | - André Mégarbané
- Department of Human Genetics, Gilbert and RoseMary Chagoury Hospital, Lebanese American University, Byblos, Lebanon
| | - Marc Bartoli
- Aix Marseille Univ, Inserm, MMG, U 1251, Institut Marseille Maladies Rares (MarMaRa), Marseille, France
| | - Frédéric Brocard
- Aix Marseille Univ, CNRS, Institut de Neurosciences de la Timone (INT), UMR 7289, Marseille, France
| | - Valérie Delague
- Aix Marseille Univ, Inserm, MMG, U 1251, Institut Marseille Maladies Rares (MarMaRa), Marseille, France.
| |
Collapse
|
28
|
Garone MG, D'Antoni C, Rosa A. Culture of Human iPSC-Derived Motoneurons in Compartmentalized Microfluidic Devices and Quantitative Assays for Studying Axonal Phenotypes. Methods Mol Biol 2022; 2429:189-199. [PMID: 35507162 DOI: 10.1007/978-1-0716-1979-7_12] [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] [Indexed: 06/14/2023]
Abstract
In order to use induced Pluripotent Stem Cells (iPSCs) to model neurodegenerative diseases, efficient and homogeneous generation of neurons in vitro represents a key step. Here we describe a method to obtain and characterize functional human spinal and cranial motoneurons using a combined approach of microfluidic chips and programs designed for scientific multidimensional imaging. We have used this approach to analyze axonal phenotypes. These tools are useful to investigate the cellular and molecular bases of neuromuscular diseases, including amyotrophic lateral sclerosis and spinal muscular atrophy.
Collapse
Affiliation(s)
- Maria Giovanna Garone
- Department of Biology and Biotechnologies "Charles Darwin", Sapienza University of Rome, Rome, Italy
| | - Chiara D'Antoni
- Department of Biology and Biotechnologies "Charles Darwin", Sapienza University of Rome, Rome, Italy
- Laboratory Affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Department of Biology and Biotechnologies "Charles Darwin", Sapienza University of Rome, Rome, Italy
| | - Alessandro Rosa
- Department of Biology and Biotechnologies "Charles Darwin", Sapienza University of Rome, Rome, Italy.
- Laboratory Affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Department of Biology and Biotechnologies "Charles Darwin", Sapienza University of Rome, Rome, Italy.
- Center for Life Nano- & Neuro-Science, Fondazione Istituto Italiano di Tecnologia, Rome, Italy.
| |
Collapse
|
29
|
OUP accepted manuscript. Stem Cells 2022; 40:175-189. [DOI: 10.1093/stmcls/sxab014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Accepted: 10/28/2021] [Indexed: 11/14/2022]
|
30
|
Zhang GY, Lv ZM, Ma HX, Chen Y, Yuan Y, Sun PX, Feng YQ, Li YW, Lu WJ, Yang YD, Yang C, Yu XL, Wang C, Liang SL, Zhang ML, Li HL, Li WL. Chemical approach to generating long-term self-renewing pMN progenitors from human embryonic stem cells. J Mol Cell Biol 2021; 14:6459209. [PMID: 34893854 PMCID: PMC8872822 DOI: 10.1093/jmcb/mjab076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 10/24/2021] [Accepted: 12/06/2021] [Indexed: 11/13/2022] Open
Abstract
Spinal cord impairment involving motor neuron degeneration and demyelination can cause life-long disabilities, but effective clinical interventions for restoring neurological functions have yet been developed. In early spinal cord development, neural progenitors in the pMN ('progenitors of motor neurons') domain, defined by the expression of oligodendrocyte transcription factor 2 (OLIG2), in ventral spinal cord first generate motor neurons and then switch the fate to produce myelin-forming oligodendrocytes. Given their differentiation potential, pMN progenitors could be a valuable cell source for cell therapy in relevant neurological conditions such as spinal cord injury. However, fast generation and expansion of pMN progenitors in vitro while conserving their differentiation potential has so far been technically challenging. In this study, based on the chemical screening, we have developed a new recipe for efficient induction of pMN progenitors from human embryonic stem cells. More importantly, these OLIG2+ pMN progenitors can be stably maintained for multiple passages without losing their ability to produce spinal motor neurons and oligodendrocytes rapidly. Our results suggest that these self-renewing pMN progenitors could potentially be useful as a renewable source of cell transplants for spinal cord injury and demyelinating disorders.
Collapse
Affiliation(s)
- Guan-Yu Zhang
- Department of Cell Biology, Second Military Medical University, Shanghai 200433, China
| | - Zhu-Man Lv
- Department of Cell Biology, Second Military Medical University, Shanghai 200433, China
| | - Hao-Xin Ma
- Department of Cell Biology, Second Military Medical University, Shanghai 200433, China
| | - Yu Chen
- Changhai Hospital, Second Military Medical University, Shanghai 200433, China
| | - Yuan Yuan
- Department of Cell Biology, Second Military Medical University, Shanghai 200433, China
| | - Ping-Xin Sun
- Department of Cell Biology, Second Military Medical University, Shanghai 200433, China
| | - Yu-Qi Feng
- Department of Cell Biology, Second Military Medical University, Shanghai 200433, China
| | - Ya-Wen Li
- Department of Histoembryology, Genetics and Developmental Biology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China.,Shanghai Key Laboratory of Reproductive Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Wen-Jie Lu
- Department of Histoembryology, Genetics and Developmental Biology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China.,Shanghai Key Laboratory of Reproductive Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Yu-Dong Yang
- Department of Histoembryology, Genetics and Developmental Biology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China.,Shanghai Key Laboratory of Reproductive Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Cheng Yang
- Wolfson Institute for Biomedical Research, University College London, Gower Street, London WC1E 6BT, UK
| | - Xin-Lu Yu
- Department of Cell Biology, Second Military Medical University, Shanghai 200433, China
| | - Chao Wang
- Department of Cell Biology, Second Military Medical University, Shanghai 200433, China
| | - Shu-Long Liang
- Department of Cell Biology, Second Military Medical University, Shanghai 200433, China
| | - Ming-Liang Zhang
- Department of Histoembryology, Genetics and Developmental Biology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China.,Shanghai Key Laboratory of Reproductive Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Hui-Liang Li
- Wolfson Institute for Biomedical Research, University College London, Gower Street, London WC1E 6BT, UK
| | - Wen-Lin Li
- Department of Cell Biology, Second Military Medical University, Shanghai 200433, China.,Shanghai Key Laboratory of Cell Engineering, Second Military Medical University, Shanghai 200433, China
| |
Collapse
|
31
|
Zheng Y, Zhang F, Xu S, Wu L. Advances in neural organoid systems and their application in neurotoxicity testing of environmental chemicals. Genes Environ 2021; 43:39. [PMID: 34551827 PMCID: PMC8456188 DOI: 10.1186/s41021-021-00214-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Accepted: 09/05/2021] [Indexed: 12/15/2022] Open
Abstract
Due to the complex structure and function of central nervous system (CNS), human CNS in vitro modeling is still a great challenge. Neurotoxicity testing of environmental chemicals mainly depends on the traditional animal models, which have various limitations such as species differences, expensive and time-consuming. Meanwhile, in vitro two-dimensional (2D) cultured cells or three-dimensional (3D) cultured neurospheres cannot fully simulate complex 3D structure of neural tissues. Recent advancements in neural organoid systems provides excellent models for the testing of environmental chemicals that affect the development of human CNS. Neural organoids derived from hPSCs not only can simulate the process of CNS development, including early stage neural tube formation, neuroepithelium differentiation and regional specification, but also its 3D structure, thus can be used to evaluate the effect of chemicals on differentiation and morphogenesis. Here, we provide a review of recent progress in the methods of culturing neural organoids and their applications in neurotoxicity testing of environmental chemicals. We conclude by highlighting challenge and future directions in neurotoxicity testing based on neural organoids.
Collapse
Affiliation(s)
- Yuanyuan Zheng
- Information Materials and Intelligent Sensing Laboratory of Anhui Province, Institutes of Physical Science and Information Technology, Anhui University, Hefei, 230601, China
| | - Fangrong Zhang
- Information Materials and Intelligent Sensing Laboratory of Anhui Province, Institutes of Physical Science and Information Technology, Anhui University, Hefei, 230601, China
| | - Shengmin Xu
- Information Materials and Intelligent Sensing Laboratory of Anhui Province, Institutes of Physical Science and Information Technology, Anhui University, Hefei, 230601, China
| | - Lijun Wu
- Information Materials and Intelligent Sensing Laboratory of Anhui Province, Institutes of Physical Science and Information Technology, Anhui University, Hefei, 230601, China.
| |
Collapse
|
32
|
Kato Y, Sakamoto K. Niclosamide affects intracellular TDP-43 distribution in motor neurons, activates mitophagy, and attenuates morphological changes under stress. J Biosci Bioeng 2021; 132:640-650. [PMID: 34429248 DOI: 10.1016/j.jbiosc.2021.06.015] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2021] [Revised: 06/29/2021] [Accepted: 06/30/2021] [Indexed: 12/15/2022]
Abstract
Amyotrophic lateral sclerosis (ALS) is characterized by progressive motor neuron loss in the brain and spinal cord; however, its etiology is unknown, and no curative treatment exists. TAR DNA-binding protein 43 (TDP-43), encoded by TARDBP, is a genetic mutation observed in 2-5% of familial ALS, and TDP is known to be mislocalized in the cytoplasm. This study aimed to identify compounds that inhibited the nuclear to cytoplasmic localization of TDP-43 in human induced pluripotent stem (iPS) cells-derived neurons. TDP-43 transgenic human iPS cells were constructed, differentiated into motor neurons, and then treated with MG-132 and sodium arsenite (stressors) to induce nuclear to cytoplasmic localization of TDP-43. STAT3 inhibitors, such as niclosamide, prevented TDP-43 mislocalization and degraded TDP-43 aggregates, and attenuated morphological changes under stress. Furthermore, niclosamide activated mitophagy via the PINK1-parkin-ubiquitin pathway. These findings suggest niclosamide may be a therapeutic candidate for ALS.
Collapse
Affiliation(s)
- Yosuke Kato
- Graduate School of Life and Environmental Science, University of Tsukuba, 1-1-1 Tennoudai, Tsukuba, Ibaraki 305-8572, Japan; Discovery Technology Laboratories, Sohyaku, Innovative Research Division, Mitsubishi Tanabe Pharma Corporation, 2-26-1 Muraoka-Higashi, Fujisawa, Kanagawa 251-8555, Japan
| | - Kazuichi Sakamoto
- Graduate School of Life and Environmental Science, University of Tsukuba, 1-1-1 Tennoudai, Tsukuba, Ibaraki 305-8572, Japan.
| |
Collapse
|
33
|
Olmsted ZT, Stigliano C, Scimemi A, Wolfe T, Cibelli J, Horner PJ, Paluh JL. Transplantable human motor networks as a neuron-directed strategy for spinal cord injury. iScience 2021; 24:102827. [PMID: 34381965 PMCID: PMC8333163 DOI: 10.1016/j.isci.2021.102827] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Revised: 05/03/2021] [Accepted: 07/05/2021] [Indexed: 12/12/2022] Open
Abstract
To repair neural circuitry following spinal cord injury (SCI), neural stem cell (NSC) transplantation has held a primary focus; however, stochastic outcomes generate challenges driven in part by NSC differentiation and tumor formation. The recent ability to generate regionally specific neurons and their support cells now allows consideration of directed therapeutic approaches with pre-differentiated and networked spinal neural cells. Here, we form encapsulated, transplantable neuronal networks of regionally matched cervical spinal motor neurons, interneurons, and oligodendrocyte progenitor cells derived through trunk-biased neuromesodermal progenitors. We direct neurite formation in alginate-based neural ribbons to generate electrically active, synaptically connected networks, characterized by electrophysiology and calcium imaging before transplantation into rodent models of contused SCI for evaluation at 10-day and 6-week timepoints. The in vivo analyses demonstrate viability and retention of interconnected synaptic networks that readily integrate with the host parenchyma to advance goals of transplantable neural circuitry for SCI treatment. Neuromesodermal progenitor derivation of human spinal neurons as therapeutic cells Neural ribbons bridge in vitro network formation and in vivo host transplantation In vivo visualization of encapsulated graft placement with magnetic resonance imaging Six-week viability of human neuronal networks with OPCs in rat contusion SCI
Collapse
Affiliation(s)
- Zachary T. Olmsted
- State University of New York Polytechnic Institute, College of Nanoscale Science and Engineering, Nanobioscience Constellation, 257 Fuller Road, Albany, NY 12203, USA
| | - Cinzia Stigliano
- Houston Methodist Research Institute, Department of Neurosurgery, Center for Neuroregeneration, 6670 Bertner Avenue R10-North, Houston, TX 77030, USA
| | - Annalisa Scimemi
- State University of New York at Albany, Biological Sciences, 1400 Washington Avenue, Albany, NY 12222, USA
| | - Tatiana Wolfe
- Houston Methodist Research Institute, Department of Neurosurgery, Center for Neuroregeneration, 6670 Bertner Avenue R10-North, Houston, TX 77030, USA
| | - Jose Cibelli
- Michigan State University, Department of Animal Science, College of Agriculture and Natural Resources and Large Animal Clinical Sciences, College of Veterinary Medicine, East Lansing, MI48824, USA
| | - Philip J. Horner
- Houston Methodist Research Institute, Department of Neurosurgery, Center for Neuroregeneration, 6670 Bertner Avenue R10-North, Houston, TX 77030, USA
| | - Janet L. Paluh
- State University of New York Polytechnic Institute, College of Nanoscale Science and Engineering, Nanobioscience Constellation, 257 Fuller Road, Albany, NY 12203, USA
- Corresponding author
| |
Collapse
|
34
|
Rayon T, Briscoe J. Cross-species comparisons and in vitro models to study tempo in development and homeostasis. Interface Focus 2021; 11:20200069. [PMID: 34055305 PMCID: PMC8086913 DOI: 10.1098/rsfs.2020.0069] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/22/2021] [Indexed: 12/14/2022] Open
Abstract
Time is inherent to biological processes. It determines the order of events and the speed at which they take place. However, we still need to refine approaches to measure the course of time in biological systems and understand what controls the pace of development. Here, we argue that the comparison of biological processes across species provides molecular insight into the timekeeping mechanisms in biology. We discuss recent findings and the open questions in the field and highlight the use of in vitro systems as tools to investigate cell-autonomous control as well as the coordination of temporal mechanisms within tissues. Further, we discuss the relevance of studying tempo for tissue transplantation, homeostasis and lifespan.
Collapse
|
35
|
Busby L, Steventon B. Tissue tectonics and the multi-scale regulation of developmental timing. Interface Focus 2021; 11:20200057. [PMID: 34055304 PMCID: PMC8086930 DOI: 10.1098/rsfs.2020.0057] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/22/2021] [Indexed: 12/17/2022] Open
Abstract
Development encompasses processes that occur at multiple length scales, including gene-regulatory interactions, cell movements and reorganization, cell signalling and growth. It is essential that the timing of events in all of these different processes is coordinated to generate well-patterned tissues and organs. However, how the timing of intrinsic cell state changes is coordinated with events occurring at the multi-tissue and whole-organism level is unknown. Here, we argue that an important mechanism that accounts for the integration of timing across levels of organization is provided by tissue tectonics, i.e. how morphogenetic events driving tissue shape changes result in the relative displacement of signalling and responding tissues and coordinate developmental timing across scales. In doing so, tissue tectonics provides a mechanism by which the cell specification events intrinsic to cells can be modulated by the temporal exposure to extracellular signals. This exposure is in turn regulated by higher-order properties of the embryo, such as their physical properties, rates of growth and the combination of dynamic cell behaviours, impacting tissue morphogenesis. Tissue tectonics creates a downward flow of information from higher to lower levels of biological organization, providing an instance of downward causation in development.
Collapse
Affiliation(s)
- Lara Busby
- Department of Genetics, University of Cambridge, Cambridge CB2 3EH, UK
| | | |
Collapse
|
36
|
Zhao A, Pan Y, Cai S. Patient-Specific Cells for Modeling and Decoding Amyotrophic Lateral Sclerosis: Advances and Challenges. Stem Cell Rev Rep 2021; 16:482-502. [PMID: 31916190 DOI: 10.1007/s12015-019-09946-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Motor neuron loss or degeneration is the typical characteristic of amyotrophic lateral sclerosis (ALS), which often leads to weakness, paralysis, or even death. The underlying mechanisms of motor neuron degeneration and ALS progression remain elusive, and there is no effective treatment for ALS. The advances of stem cells and reprogramming techniques has made it possible to generate patient-specific motor neurons as cell models for studying disease mechanisms and drug discovery. This review comprehensively discusses recent approaches to generate motor neurons from stem cells and somatic cells and highlights the application of induced motor neurons to modeling ALS diseases, dissecting the pathogenesis, and screening new drugs. New perspectives are also discussed on generating patient-specific motor neuron subtypes that are affected by ALS or creating 3D spinal cord organoid models for better recapitulating and understanding ALS.
Collapse
Affiliation(s)
- Andong Zhao
- Health Science Center, Shenzhen University, Shenzhen, 518060, China
| | - Yu Pan
- Department of Trauma and Orthopedics, The 2nd Affiliated Hospital of Shenzhen University, The Affiliated Baoan Hospital of Southern Medical University, Shenzhen, 518101, China.
| | - Sa Cai
- Health Science Center, Shenzhen University, Shenzhen, 518060, China.
| |
Collapse
|
37
|
Cutarelli A, Martínez-Rojas VA, Tata A, Battistella I, Rossi D, Arosio D, Musio C, Conti L. A Monolayer System for the Efficient Generation of Motor Neuron Progenitors and Functional Motor Neurons from Human Pluripotent Stem Cells. Cells 2021; 10:cells10051127. [PMID: 34066970 PMCID: PMC8151197 DOI: 10.3390/cells10051127] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 04/27/2021] [Accepted: 05/04/2021] [Indexed: 12/12/2022] Open
Abstract
Methods for the conversion of human induced pluripotent stem cells (hiPSCs) into motor neurons (MNs) have opened to the generation of patient-derived in vitro systems that can be exploited for MN disease modelling. However, the lack of simplified and consistent protocols and the fact that hiPSC-derived MNs are often functionally immature yet limit the opportunity to fully take advantage of this technology, especially in research aimed at revealing the disease phenotypes that are manifested in functionally mature cells. In this study, we present a robust, optimized monolayer procedure to rapidly convert hiPSCs into enriched populations of motor neuron progenitor cells (MNPCs) that can be further amplified to produce a large number of cells to cover many experimental needs. These MNPCs can be efficiently differentiated towards mature MNs exhibiting functional electrical and pharmacological neuronal properties. Finally, we report that MN cultures can be long-term maintained, thus offering the opportunity to study degenerative phenomena associated with pathologies involving MNs and their functional, networked activity. These results indicate that our optimized procedure enables the efficient and robust generation of large quantities of MNPCs and functional MNs, providing a valid tool for MNs disease modelling and for drug discovery applications.
Collapse
Affiliation(s)
- Alessandro Cutarelli
- Laboratory of Stem Cell Biology, Department of Cellular, Computational and Integrative Biology-CIBIO, University of Trento, 38123 Trento, Italy; (A.C.); (A.T.); (I.B.)
| | - Vladimir A. Martínez-Rojas
- Institute of Biophysics (IBF), Trento Unit, National Research Council (CNR) & LabSSAH, Bruno Kessler Foundation (FBK), 38123 Trento, Italy; (V.A.M.-R.); (D.A.); (C.M.)
| | - Alice Tata
- Laboratory of Stem Cell Biology, Department of Cellular, Computational and Integrative Biology-CIBIO, University of Trento, 38123 Trento, Italy; (A.C.); (A.T.); (I.B.)
| | - Ingrid Battistella
- Laboratory of Stem Cell Biology, Department of Cellular, Computational and Integrative Biology-CIBIO, University of Trento, 38123 Trento, Italy; (A.C.); (A.T.); (I.B.)
| | - Daniela Rossi
- Laboratory for Research on Neurodegenerative Disorders, Istituti Clinici Scientifici Maugeri IRCCS, 27100 Pavia, Italy;
| | - Daniele Arosio
- Institute of Biophysics (IBF), Trento Unit, National Research Council (CNR) & LabSSAH, Bruno Kessler Foundation (FBK), 38123 Trento, Italy; (V.A.M.-R.); (D.A.); (C.M.)
| | - Carlo Musio
- Institute of Biophysics (IBF), Trento Unit, National Research Council (CNR) & LabSSAH, Bruno Kessler Foundation (FBK), 38123 Trento, Italy; (V.A.M.-R.); (D.A.); (C.M.)
| | - Luciano Conti
- Laboratory of Stem Cell Biology, Department of Cellular, Computational and Integrative Biology-CIBIO, University of Trento, 38123 Trento, Italy; (A.C.); (A.T.); (I.B.)
- Correspondence: ; Tel.: +39-0461-285216
| |
Collapse
|
38
|
McIntyre WB, Pieczonka K, Khazaei M, Fehlings MG. Regenerative replacement of neural cells for treatment of spinal cord injury. Expert Opin Biol Ther 2021; 21:1411-1427. [PMID: 33830863 DOI: 10.1080/14712598.2021.1914582] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Introduction: Traumatic Spinal Cord Injury (SCI) results from primary physical injury to the spinal cord, which initiates a secondary cascade of neural cell death. Current therapeutic approaches can attenuate the consequences of the primary and secondary events, but do not address the degenerative aspects of SCI. Transplantation of neural stem/progenitor cells (NPCs) for the replacement of the lost/damaged neural cells is suggested here as a regenerative approach that is complementary to current therapeutics.Areas Covered: This review addresses how neurons, oligodendrocytes, and astrocytes are impacted by traumatic SCI, and how current research in regenerative-NPC therapeutics aims to restore their functionality. Methods used to enhance graft survival, as well as bias progenitor cells towards neuronal, oligodendrogenic, and astroglia lineages are discussed.Expert Opinion: Despite an NPC's ability to differentiate into neurons, oligodendrocytes, and astrocytes in the transplant environment, their potential therapeutic efficacy requires further optimization prior to translation into the clinic. Considering the temporospatial identity of NPCs could promote neural repair in region specific injuries throughout the spinal cord. Moreover, understanding which cells are targeted by NPC-derived myelinating cells can help restore physiologically-relevant myelin patterns. Finally, the duality of astrocytes is discussed, outlining their context-dependent importance in the treatment of SCI.
Collapse
Affiliation(s)
- William Brett McIntyre
- Division of Genetics and Development, Krembil Research Institute, University Health Network, Toronto, ON, Canada.,Institute of Medical Sciences, University of Toronto, Toronto, ON, Canada
| | - Katarzyna Pieczonka
- Division of Genetics and Development, Krembil Research Institute, University Health Network, Toronto, ON, Canada.,Institute of Medical Sciences, University of Toronto, Toronto, ON, Canada
| | - Mohamad Khazaei
- Division of Genetics and Development, Krembil Research Institute, University Health Network, Toronto, ON, Canada
| | - Michael G Fehlings
- Division of Genetics and Development, Krembil Research Institute, University Health Network, Toronto, ON, Canada.,Institute of Medical Sciences, University of Toronto, Toronto, ON, Canada.,Department of Surgery, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
39
|
Olmsted ZT, Paluh JL. Stem Cell Neurodevelopmental Solutions for Restorative Treatments of the Human Trunk and Spine. Front Cell Neurosci 2021; 15:667590. [PMID: 33981202 PMCID: PMC8107236 DOI: 10.3389/fncel.2021.667590] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2021] [Accepted: 03/29/2021] [Indexed: 12/21/2022] Open
Abstract
The ability to reliably repair spinal cord injuries (SCI) will be one of the greatest human achievements realized in regenerative medicine. Until recently, the cellular path to this goal has been challenging. However, as detailed developmental principles are revealed in mouse and human models, their application in the stem cell community brings trunk and spine embryology into efforts to advance human regenerative medicine. New models of posterior embryo development identify neuromesodermal progenitors (NMPs) as a major bifurcation point in generating the spinal cord and somites and is leading to production of cell types with the full range of axial identities critical for repair of trunk and spine disorders. This is coupled with organoid technologies including assembloids, circuitoids, and gastruloids. We describe a paradigm for applying developmental principles towards the goal of cell-based restorative therapies to enable reproducible and effective near-term clinical interventions.
Collapse
|
40
|
Wind M, Gogolou A, Manipur I, Granata I, Butler L, Andrews PW, Barbaric I, Ning K, Guarracino MR, Placzek M, Tsakiridis A. Defining the signalling determinants of a posterior ventral spinal cord identity in human neuromesodermal progenitor derivatives. Development 2021; 148:dev194415. [PMID: 33658223 PMCID: PMC8015249 DOI: 10.1242/dev.194415] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Accepted: 02/23/2021] [Indexed: 12/14/2022]
Abstract
The anteroposterior axial identity of motor neurons (MNs) determines their functionality and vulnerability to neurodegeneration. Thus, it is a crucial parameter in the design of strategies aiming to produce MNs from human pluripotent stem cells (hPSCs) for regenerative medicine/disease modelling applications. However, the in vitro generation of posterior MNs corresponding to the thoracic/lumbosacral spinal cord has been challenging. Although the induction of cells resembling neuromesodermal progenitors (NMPs), the bona fide precursors of the spinal cord, offers a promising solution, the progressive specification of posterior MNs from these cells is not well defined. Here, we determine the signals guiding the transition of human NMP-like cells toward thoracic ventral spinal cord neurectoderm. We show that combined WNT-FGF activities drive a posterior dorsal pre-/early neural state, whereas suppression of TGFβ-BMP signalling pathways promotes a ventral identity and neural commitment. Based on these results, we define an optimised protocol for the generation of thoracic MNs that can efficiently integrate within the neural tube of chick embryos. We expect that our findings will facilitate the comparison of hPSC-derived spinal cord cells of distinct axial identities.
Collapse
Affiliation(s)
- Matthew Wind
- Centre for Stem Cell Biology, Department of Biomedical Science, The University of Sheffield, Sheffield S10 2TN, UK
- Department of Biomedical Science and Bateson Centre, University of Sheffield, Sheffield S10 2TN, UK
- Department of Neuroscience, Neuroscience Institute, University of Sheffield, Western Bank, Sheffield S10 2TN, UK
| | - Antigoni Gogolou
- Centre for Stem Cell Biology, Department of Biomedical Science, The University of Sheffield, Sheffield S10 2TN, UK
- Department of Biomedical Science and Bateson Centre, University of Sheffield, Sheffield S10 2TN, UK
- Department of Neuroscience, Neuroscience Institute, University of Sheffield, Western Bank, Sheffield S10 2TN, UK
| | - Ichcha Manipur
- Computational and Data Science Laboratory, High Performance Computing and Networking Institute, National Research Council of Italy, Napoli 80131, Italy
| | - Ilaria Granata
- Computational and Data Science Laboratory, High Performance Computing and Networking Institute, National Research Council of Italy, Napoli 80131, Italy
| | - Larissa Butler
- Centre for Stem Cell Biology, Department of Biomedical Science, The University of Sheffield, Sheffield S10 2TN, UK
| | - Peter W Andrews
- Centre for Stem Cell Biology, Department of Biomedical Science, The University of Sheffield, Sheffield S10 2TN, UK
| | - Ivana Barbaric
- Centre for Stem Cell Biology, Department of Biomedical Science, The University of Sheffield, Sheffield S10 2TN, UK
| | - Ke Ning
- Department of Neuroscience, Neuroscience Institute, University of Sheffield, Western Bank, Sheffield S10 2TN, UK
- Sheffield Institute for Translational Neuroscience, Department of Neuroscience, University of Sheffield, Sheffield S10 2HQ, UK
| | | | - Marysia Placzek
- Department of Biomedical Science and Bateson Centre, University of Sheffield, Sheffield S10 2TN, UK
| | - Anestis Tsakiridis
- Centre for Stem Cell Biology, Department of Biomedical Science, The University of Sheffield, Sheffield S10 2TN, UK
- Department of Biomedical Science and Bateson Centre, University of Sheffield, Sheffield S10 2TN, UK
- Department of Neuroscience, Neuroscience Institute, University of Sheffield, Western Bank, Sheffield S10 2TN, UK
| |
Collapse
|
41
|
Vieira de Sá R, Cañizares Luna M, Pasterkamp RJ. Advances in Central Nervous System Organoids: A Focus on Organoid-Based Models for Motor Neuron Disease. Tissue Eng Part C Methods 2021; 27:213-224. [PMID: 33446055 DOI: 10.1089/ten.tec.2020.0337] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Despite their large societal burden, the development of therapeutic treatments for neurodegenerative diseases (NDDs) has been relatively unsuccessful. This is, in part, due to a lack of representative experimental models that reveal fundamental aspects of human brain pathology. Recently, assays for in vitro modeling of the human central nervous system (CNS) have significantly improved with the development of brain and spinal cord organoids. Coupled with induced-pluripotent stem cell and genome editing technologies, CNS organoids are a promising tool for studying neurodegeneration in a patient-specific manner. An extensive array of protocols for the generation of organoids for different brain regions has been developed and used for studying neurodegenerative and other brain diseases. However, their application in the field of motor neuron disease (MND) has been limited due to a lack of adequate organoid models. The development of protocols to derive spinal cord and trunk organoids and progress in the field of assembloids are providing new opportunities for modeling MND. In this study here we review recent advances in the development of CNS organoid models, their application in NDDs, and technical limitations. Finally, we discuss future perspectives for the development of organoid-based systems for MND and provide a framework for their development. Impact statement Animal models and two-dimensional cultures are currently the main platforms for studying neurodegenerative diseases (NDDs). However, central nervous system (CNS) organoid technology offers novel possibilities for studying these diseases. Organoid modeling in combination with emerging organ-on-a-chip approaches, induced-pluripotent stem cell technology, and genome editing render in vitro modeling of NDDs more robust and physiologically relevant. In this study, we review the principles underlying CNS organoid generation, their use in NDD research, and future perspectives in organoid technology. Finally, we discuss how advances in different fields could be combined to generate a multisystem organoid-on-a-chip model to investigate a specific class of NDDs, motor neuron diseases.
Collapse
Affiliation(s)
- Renata Vieira de Sá
- Department of Translational Neuroscience, University Medical Center Utrecht Brain Center, Utrecht University, Utrecht, The Netherlands
| | - Marta Cañizares Luna
- Department of Translational Neuroscience, University Medical Center Utrecht Brain Center, Utrecht University, Utrecht, The Netherlands
| | - R Jeroen Pasterkamp
- Department of Translational Neuroscience, University Medical Center Utrecht Brain Center, Utrecht University, Utrecht, The Netherlands
| |
Collapse
|
42
|
Abstract
When nerves are damaged by trauma or disease, they are still capable of firing off electrical command signals that originate from the brain. Furthermore, those damaged nerves have an innate ability to partially regenerate, so they can heal from trauma and even reinnervate new muscle targets. For an amputee who has his/her damaged nerves surgically reconstructed, the electrical signals that are generated by the reinnervated muscle tissue can be sensed and interpreted with bioelectronics to control assistive devices or robotic prostheses. No two amputees will have identical physiologies because there are many surgical options for reconstructing residual limbs, which may in turn impact how well someone can interface with a robotic prosthesis later on. In this review, we aim to investigate what the literature has to say about different pathways for peripheral nerve regeneration and how each pathway can impact the neuromuscular tissue’s final electrophysiology. This information is important because it can guide us in planning the development of future bioelectronic devices, such as prosthetic limbs or neurostimulators. Future devices will primarily have to interface with tissue that has undergone some natural regeneration process, and so we have explored and reported here what is known about the bioelectrical features of neuromuscular tissue regeneration.
Collapse
|
43
|
de Leeuw VC, Pennings JLA, Hessel EVS, Piersma AH. Exploring the biological domain of the neural embryonic stem cell test (ESTn): Morphogenetic regulators, Hox genes and cell types, and their usefulness as biomarkers for embryotoxicity screening. Toxicology 2021; 454:152735. [PMID: 33636252 DOI: 10.1016/j.tox.2021.152735] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Revised: 01/25/2021] [Accepted: 02/20/2021] [Indexed: 11/25/2022]
Abstract
Animal-free assessment of compound-induced developmental neurotoxicity will most likely be based on batteries of multiple in vitro tests. The optimal battery is built by combining tests with complementary biological domains that together ideally cover all relevant toxicity pathways. Thus, biological domain definition, i.e. which biological processes and cell types are represented, is an important assay characteristic for determining the place of assays in testing strategies. The murine neural embryonic stem cell test (ESTn) is employed to predict the developmental neurotoxicity of compounds. The aim of this study was to explore the biological domain of ESTn according to three groups of biomarker genes of early (neuro)development: morphogenetic regulators, Hox genes and cell type markers for the ectodermal and neural lineages. These biomarker groups were selected based on their crucial regulatory role in (neuro)development. Analysis of these genes in a series of previously generated whole transcriptome datasets of ESTn showed that at day 7 in culture cell differentiation resembled hindbrain/branchial/thoracic development between E6.5-E12.5 in vivo, with subsequent development into a mixed cell culture containing different neural subtypes, astrocytes and oligodendrocytes by day 13. In addition, the selected biomarkers showed common and distinct responses to compound exposure. Monitoring the biological domain of ESTn through gene expression patterns of morphogenetic regulators, Hox genes and cell type markers proved instrumental in providing mechanistic understanding of compound effects on neural differentiation in ESTn, and can aid in positioning of the test in a battery of complementary in vitro tests in integrated approaches to testing and assessment.
Collapse
Affiliation(s)
- Victoria C de Leeuw
- Centre for Health Protection, National Institute for Public Health and the Environment (RIVM), Bilthoven, the Netherlands; Institute for Risk Assessment Sciences (IRAS), Utrecht University, Utrecht, the Netherlands.
| | - Jeroen L A Pennings
- Centre for Health Protection, National Institute for Public Health and the Environment (RIVM), Bilthoven, the Netherlands
| | - Ellen V S Hessel
- Centre for Health Protection, National Institute for Public Health and the Environment (RIVM), Bilthoven, the Netherlands
| | - Aldert H Piersma
- Centre for Health Protection, National Institute for Public Health and the Environment (RIVM), Bilthoven, the Netherlands; Institute for Risk Assessment Sciences (IRAS), Utrecht University, Utrecht, the Netherlands
| |
Collapse
|
44
|
Solomon E, Davis-Anderson K, Hovde B, Micheva-Viteva S, Harris JF, Twary S, Iyer R. Global transcriptome profile of the developmental principles of in vitro iPSC-to-motor neuron differentiation. BMC Mol Cell Biol 2021; 22:13. [PMID: 33602141 PMCID: PMC7893891 DOI: 10.1186/s12860-021-00343-z] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Accepted: 01/11/2021] [Indexed: 12/30/2022] Open
Abstract
Background Human induced pluripotent stem cells (iPSC) have opened new avenues for regenerative medicine. Consequently, iPSC-derived motor neurons have emerged as potentially viable therapies for spinal cord injuries and neurodegenerative disorders including Amyotrophic Lateral Sclerosis. However, direct clinical application of iPSC bears in itself the risk of tumorigenesis and other unforeseeable genetic or epigenetic abnormalities. Results Employing RNA-seq technology, we identified and characterized gene regulatory networks triggered by in vitro chemical reprogramming of iPSC into cells with the molecular features of motor neurons (MNs) whose function in vivo is to innervate effector organs. We present meta-transcriptome signatures of 5 cell types: iPSCs, neural stem cells, motor neuron progenitors, early motor neurons, and mature motor neurons. In strict response to the chemical stimuli, along the MN differentiation axis we observed temporal downregulation of tumor growth factor-β signaling pathway and consistent activation of sonic hedgehog, Wnt/β-catenin, and Notch signaling. Together with gene networks defining neuronal differentiation (neurogenin 2, microtubule-associated protein 2, Pax6, and neuropilin-1), we observed steady accumulation of motor neuron-specific regulatory genes, including Islet-1 and homeobox protein HB9. Interestingly, transcriptome profiling of the differentiation process showed that Ca2+ signaling through cAMP and LPC was downregulated during the conversion of the iPSC to neural stem cells and key regulatory gene activity of the pathway remained inhibited until later stages of motor neuron formation. Pathways shaping the neuronal development and function were well-represented in the early motor neuron cells including, neuroactive ligand-receptor interactions, axon guidance, and the cholinergic synapse formation. A notable hallmark of our in vitro motor neuron maturation in monoculture was the activation of genes encoding G-coupled muscarinic acetylcholine receptors and downregulation of the ionotropic nicotinic acetylcholine receptors expression. We observed the formation of functional neuronal networks as spontaneous oscillations in the extracellular action potentials recorded on multi-electrode array chip after 20 days of differentiation. Conclusions Detailed transcriptome profile of each developmental step from iPSC to motor neuron driven by chemical induction provides the guidelines to novel therapeutic approaches in the re-construction efforts of muscle innervation. Supplementary Information The online version contains supplementary material available at 10.1186/s12860-021-00343-z.
Collapse
Affiliation(s)
- Emilia Solomon
- Los Alamos National Laboratory, Bioscience Division, Los Alamos, NM, USA
| | | | - Blake Hovde
- Los Alamos National Laboratory, Bioscience Division, Los Alamos, NM, USA
| | | | | | - Scott Twary
- Los Alamos National Laboratory, Bioscience Division, Los Alamos, NM, USA
| | - Rashi Iyer
- Los Alamos National Laboratory, Analytics, Intelligence, and Technology Division, Los Alamos, NM, USA.
| |
Collapse
|
45
|
Otake K, Adachi-Tominari K, Nagai H, Saito M, Sano O, Hirozane Y, Iwata H. Quantitative comparison of the mRNA content of human iPSC-derived motor neurons and their extracellular vesicles. FEBS Open Bio 2021; 11:494-506. [PMID: 33296136 PMCID: PMC7876496 DOI: 10.1002/2211-5463.13059] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Revised: 11/30/2020] [Accepted: 12/07/2020] [Indexed: 01/22/2023] Open
Abstract
Extracellular vesicles (EVs) contain various cargo molecules, including RNAs and proteins. EVs, which include exosomes, are predicted to be suitable surrogates of their source cells for liquid biopsy to measure biomarkers. Several studies have performed qualitative comparisons of cargo molecule repertoires between source cells and their EVs. However, quantitative comparisons have not been reported so far. Furthermore, many studies analyzed microRNAs or proteins in EVs, but not mRNAs. In this study, we analyzed mRNAs in motor neurons and their EVs. Normal human-induced pluripotent stem cells were differentiated into motor neurons, and comprehensive analysis of mRNAs in the cells and their EVs was performed by RNA sequencing. Differential analysis between cellular and EV mRNAs was performed by edgeR after normalization of read count. The results suggest that signatures in the abundance of EV mRNAs were different from those of cellular mRNAs. Comparison of intracellular vesicle and EV mRNA abundance showed negatively and positively biased genes in the EVs. Gene Ontology analysis revealed that the genes showing negatively biased abundance in the EVs were enriched in many functions regarding neuronal development. In contrast, the positively biased genes were enriched in functions regarding cellular metabolism and protein synthesis. These results suggest that mRNAs in motor neurons are loaded into EVs to regulate certain mechanisms, which are yet to be elucidated.
Collapse
Affiliation(s)
- Kentaro Otake
- Innovative Biology Laboratories, Neuroscience Drug Discovery Unit, Research, Takeda Pharmaceutical Company Limited, Fujisawa, Japan
| | - Keiko Adachi-Tominari
- Innovative Biology Laboratories, Neuroscience Drug Discovery Unit, Research, Takeda Pharmaceutical Company Limited, Fujisawa, Japan
| | - Hiroaki Nagai
- Innovative Biology Laboratories, Neuroscience Drug Discovery Unit, Research, Takeda Pharmaceutical Company Limited, Fujisawa, Japan
| | - Masayo Saito
- Innovative Biology Laboratories, Neuroscience Drug Discovery Unit, Research, Takeda Pharmaceutical Company Limited, Fujisawa, Japan
| | - Osamu Sano
- Innovative Biology Laboratories, Neuroscience Drug Discovery Unit, Research, Takeda Pharmaceutical Company Limited, Fujisawa, Japan
| | - Yoshihiko Hirozane
- Innovative Biology Laboratories, Neuroscience Drug Discovery Unit, Research, Takeda Pharmaceutical Company Limited, Fujisawa, Japan
| | - Hidehisa Iwata
- Innovative Biology Laboratories, Neuroscience Drug Discovery Unit, Research, Takeda Pharmaceutical Company Limited, Fujisawa, Japan
| |
Collapse
|
46
|
Domínguez-Bautista JA, Acevo-Rodríguez PS, Castro-Obregón S. Programmed Cell Senescence in the Mouse Developing Spinal Cord and Notochord. Front Cell Dev Biol 2021; 9:587096. [PMID: 33575260 PMCID: PMC7870793 DOI: 10.3389/fcell.2021.587096] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Accepted: 01/05/2021] [Indexed: 01/10/2023] Open
Abstract
Programmed cell senescence is a cellular process that seems to contribute to embryo development, in addition to cell proliferation, migration, differentiation and programmed cell death, and has been observed in evolutionary distant organisms such as mammals, amphibians, birds and fish. Programmed cell senescence is a phenotype similar to stress-induced cellular senescence, characterized by the expression of the cell cycle inhibitors p21CIP1/WAF and p16INK4A, increased activity of a lysosomal enzyme with beta-galactosidase activity (coined senescence-associated beta-galactosidase) and secretion of growth factors, interleukins, chemokines, metalloproteases, etc., collectively known as a senescent-associated secretory phenotype that instructs surrounding tissue. How wide is the distribution of programmed cell senescence during mouse development and its specific mechanisms to shape the embryo are still poorly understood. Here, we investigated whether markers of programmed cell senescence are found in the developing mouse spinal cord and notochord. We found discrete areas and developmental windows with high senescence-associated beta galactosidase in both spinal cord and notochord, which was reduced in mice embryos developed ex-utero in the presence of the senolytic ABT-263. Expression of p21CIP1/WAF was documented in epithelial cells of the spinal cord and the notochord, while p16INK4A was observed in motoneurons. Treatment with the senolytic ABT-263 decreased the number of motoneurons, supporting their senescent phenotype. Our data suggest that a subpopulation of motoneurons in the developing spinal cord, as well as some notochord cells undergo programmed cell senescence.
Collapse
Affiliation(s)
| | | | - Susana Castro-Obregón
- División de Neurociencias, Instituto de Fisiología Celular, UNAM, Mexico City, Mexico
| |
Collapse
|
47
|
Bulajić M, Srivastava D, Dasen JS, Wichterle H, Mahony S, Mazzoni EO. Differential abilities to engage inaccessible chromatin diversify vertebrate Hox binding patterns. Development 2020; 147:dev194761. [PMID: 33028607 PMCID: PMC7710020 DOI: 10.1242/dev.194761] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Accepted: 09/25/2020] [Indexed: 12/17/2022]
Abstract
Although Hox genes encode for conserved transcription factors (TFs), they are further divided into anterior, central and posterior groups based on their DNA-binding domain similarity. The posterior Hox group expanded in the deuterostome clade and patterns caudal and distal structures. We aimed to address how similar Hox TFs diverge to induce different positional identities. We studied Hox TF DNA-binding and regulatory activity during an in vitro motor neuron differentiation system that recapitulates embryonic development. We found diversity in the genomic binding profiles of different Hox TFs, even among the posterior group paralogs that share similar DNA-binding domains. These differences in genomic binding were explained by differing abilities to bind to previously inaccessible sites. For example, the posterior group HOXC9 had a greater ability to bind occluded sites than the posterior HOXC10, producing different binding patterns and driving differential gene expression programs. From these results, we propose that the differential abilities of posterior Hox TFs to bind to previously inaccessible chromatin drive patterning diversification.This article has an associated 'The people behind the papers' interview.
Collapse
Affiliation(s)
- Milica Bulajić
- Department of Biology, New York University, New York, NY 10003, USA
| | - Divyanshi Srivastava
- Center for Eukaryotic Gene Regulation, Department of Biochemistry and Molecular Biology, The Pennsylvania State University, University Park, PA 16802, USA
| | - Jeremy S Dasen
- Neuroscience Institute, Department of Neuroscience and Physiology, New York University School of Medicine, New York, NY 10016, USA
| | - Hynek Wichterle
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY 10032, USA
- Department of Neuroscience, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Shaun Mahony
- Center for Eukaryotic Gene Regulation, Department of Biochemistry and Molecular Biology, The Pennsylvania State University, University Park, PA 16802, USA
| | | |
Collapse
|
48
|
Chen M, Maimaitili M, Habekost M, Gill KP, Mermet-Joret N, Nabavi S, Febbraro F, Denham M. Rapid generation of regionally specified CNS neurons by sequential patterning and conversion of human induced pluripotent stem cells. Stem Cell Res 2020; 48:101945. [DOI: 10.1016/j.scr.2020.101945] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Revised: 07/27/2020] [Accepted: 07/30/2020] [Indexed: 12/25/2022] Open
|
49
|
Rayon T, Stamataki D, Perez-Carrasco R, Garcia-Perez L, Barrington C, Melchionda M, Exelby K, Lazaro J, Tybulewicz VLJ, Fisher EMC, Briscoe J. Species-specific pace of development is associated with differences in protein stability. Science 2020; 369:eaba7667. [PMID: 32943498 PMCID: PMC7116327 DOI: 10.1126/science.aba7667] [Citation(s) in RCA: 152] [Impact Index Per Article: 30.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2020] [Accepted: 07/29/2020] [Indexed: 12/12/2022]
Abstract
Although many molecular mechanisms controlling developmental processes are evolutionarily conserved, the speed at which the embryo develops can vary substantially between species. For example, the same genetic program, comprising sequential changes in transcriptional states, governs the differentiation of motor neurons in mouse and human, but the tempo at which it operates differs between species. Using in vitro directed differentiation of embryonic stem cells to motor neurons, we show that the program runs more than twice as fast in mouse as in human. This is not due to differences in signaling, nor the genomic sequence of genes or their regulatory elements. Instead, there is an approximately two-fold increase in protein stability and cell cycle duration in human cells compared with mouse cells. This can account for the slower pace of human development and suggests that differences in protein turnover play a role in interspecies differences in developmental tempo.
Collapse
Affiliation(s)
- Teresa Rayon
- The Francis Crick Institute, London NW1 1AT, UK.
| | | | - Ruben Perez-Carrasco
- The Francis Crick Institute, London NW1 1AT, UK
- Department of Mathematics, University College London, London WC1E 6BT, UK
- Department of Life Sciences, Imperial College London, London SW7 2AZ, UK
| | | | | | | | | | | | - Victor L J Tybulewicz
- The Francis Crick Institute, London NW1 1AT, UK
- Department of Immunology and Inflammation, Imperial College, London W12 0NN, UK
| | - Elizabeth M C Fisher
- UCL Queen Square Institute of Neurology, University College London, London WC1N 3BG, UK
| | | |
Collapse
|
50
|
Olmsted ZT, Stigliano C, Badri A, Zhang F, Williams A, Koffas MAG, Xie Y, Linhardt RJ, Cibelli J, Horner PJ, Paluh JL. Fabrication of homotypic neural ribbons as a multiplex platform optimized for spinal cord delivery. Sci Rep 2020; 10:12939. [PMID: 32737387 PMCID: PMC7395100 DOI: 10.1038/s41598-020-69274-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Accepted: 07/01/2020] [Indexed: 12/13/2022] Open
Abstract
Cell therapy for the injured spinal cord will rely on combined advances in human stem cell technologies and delivery strategies. Here we encapsulate homotypic spinal cord neural stem cells (scNSCs) in an alginate-based neural ribbon delivery platform. We perform a comprehensive in vitro analysis and qualitatively demonstrate graft survival and injury site retention using a rat C4 hemi-contusion model. Pre-configured neural ribbons are transport-stable modules that enable site-ready injection, and can support scNSC survival and retention in vivo. Neural ribbons offer multifunctionality in vitro including co-encapsulation of the injury site extracellular matrix modifier chondroitinase ABC (chABC), tested here in glial scar models, and ability of cervically-patterned scNSCs to differentiate within neural ribbons and project axons for integration with 3-D external matrices. This is the first extensive in vitro characterization of neural ribbon technology, and constitutes a plausible method for reproducible delivery, placement, and retention of viable neural cells in vivo.
Collapse
Affiliation(s)
- Zachary T Olmsted
- Nanobioscience Constellation, Colleges of Nanoscale Science and Engineering, State University of New York Polytechnic Institute, NanoFab East, 257 Fuller Road, Albany, NY, 12203, USA
| | - Cinzia Stigliano
- Center for Neuroregeneration, Department of Neurosurgery, Houston Methodist Research Institute, 6670 Bertner Ave. R10-North, Houston, TX, 77030, USA
| | - Abinaya Badri
- Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, 1623 15th St, Troy, NY, 12180, USA
| | - Fuming Zhang
- Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, 1623 15th St, Troy, NY, 12180, USA
| | - Asher Williams
- Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, 1623 15th St, Troy, NY, 12180, USA
| | - Mattheos A G Koffas
- Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, 1623 15th St, Troy, NY, 12180, USA
| | - Yubing Xie
- Nanobioscience Constellation, Colleges of Nanoscale Science and Engineering, State University of New York Polytechnic Institute, NanoFab East, 257 Fuller Road, Albany, NY, 12203, USA
| | - Robert J Linhardt
- Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, 1623 15th St, Troy, NY, 12180, USA
| | - Jose Cibelli
- Department of Animal Science, College of Agriculture and Natural Resources and Large Animal Clinical Sciences, College of Veterinary Medicine, Michigan State University, East Lansing, MI, 48824, USA
| | - Philip J Horner
- Center for Neuroregeneration, Department of Neurosurgery, Houston Methodist Research Institute, 6670 Bertner Ave. R10-North, Houston, TX, 77030, USA
| | - Janet L Paluh
- Nanobioscience Constellation, Colleges of Nanoscale Science and Engineering, State University of New York Polytechnic Institute, NanoFab East, 257 Fuller Road, Albany, NY, 12203, USA.
| |
Collapse
|