1
|
Choe MS, Lo C, Park IH. Modeling forebrain regional development and connectivity by human brain organoids. Curr Opin Genet Dev 2025; 91:102324. [PMID: 39983347 DOI: 10.1016/j.gde.2025.102324] [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: 11/05/2024] [Revised: 01/26/2025] [Accepted: 02/01/2025] [Indexed: 02/23/2025]
Abstract
The forebrain is one of the most important brain structures for modern human existence, which houses the uniquely sophisticated social and cognitive functions that distinguish our species. Therefore, modeling the forebrain development by using human cells is especially critical for our understanding of the intricacies of human development and devising treatments for related diseases. Recent advancements in brain organoid fields have offered unprecedented tools to investigate forebrain development from studies on specific regions to exploring tract formation and connectivity between different regions of the forebrain. In this review, we discuss the developmental biology of the forebrain and diverse methods for modeling its development by using organoids.
Collapse
Affiliation(s)
- Mu Seog Choe
- Interdepartmental Neuroscience Program, Department of Genetics, Yale Stem Cell Center, Yale Child Study Center, Wu Tsai Institute, Yale School of Medicine, New Haven, CT, United States
| | - Cynthia Lo
- Interdepartmental Neuroscience Program, Department of Genetics, Yale Stem Cell Center, Yale Child Study Center, Wu Tsai Institute, Yale School of Medicine, New Haven, CT, United States
| | - In-Hyun Park
- Interdepartmental Neuroscience Program, Department of Genetics, Yale Stem Cell Center, Yale Child Study Center, Wu Tsai Institute, Yale School of Medicine, New Haven, CT, United States.
| |
Collapse
|
2
|
Ikeda M, Doi D, Ebise H, Ozaki Y, Fujii M, Kikuchi T, Yoshida K, Takahashi J. Validation of non-destructive morphology-based selection of cerebral cortical organoids by paired morphological and single-cell RNA-seq analyses. Stem Cell Reports 2024; 19:1635-1646. [PMID: 39393360 PMCID: PMC11589179 DOI: 10.1016/j.stemcr.2024.09.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Revised: 09/14/2024] [Accepted: 09/16/2024] [Indexed: 10/13/2024] Open
Abstract
Organoids, self-organized cell aggregates, contribute significantly to developing disease models and cell-based therapies. Organoid-to-organoid variations, however, are inevitable despite the use of the latest differentiation protocols. Here, we focused on the morphology of organoids formed in a cerebral organoid differentiation culture and assessed their cellular compositions by single-cell RNA sequencing analysis. The data revealed that organoids primarily composed of non-neuronal cells, such as those from the neural crest and choroid plexus, showed unique morphological features. Moreover, we demonstrate that non-destructive morphological analysis can accurately distinguish organoids composed of cerebral cortical tissues from other cerebral tissues, thus enhancing experimental accuracy and reliability to ensure the safety of cell-based therapies.
Collapse
Affiliation(s)
- Megumi Ikeda
- Department of Clinical Application, Center for iPS Cell Research and Application, Kyoto University, Kyoto 606-8507, Japan; Regenerative & Cellular Medicine Kobe Center, Sumitomo Pharma Co., Ltd., Chuo-ku, Kobe 650-0047, Japan
| | - Daisuke Doi
- Department of Clinical Application, Center for iPS Cell Research and Application, Kyoto University, Kyoto 606-8507, Japan
| | - Hayao Ebise
- Regenerative & Cellular Medicine Kobe Center, Sumitomo Pharma Co., Ltd., Chuo-ku, Kobe 650-0047, Japan
| | - Yuki Ozaki
- Department of Clinical Application, Center for iPS Cell Research and Application, Kyoto University, Kyoto 606-8507, Japan
| | - Misaki Fujii
- Department of Clinical Application, Center for iPS Cell Research and Application, Kyoto University, Kyoto 606-8507, Japan
| | - Tetsuhiro Kikuchi
- Department of Clinical Application, Center for iPS Cell Research and Application, Kyoto University, Kyoto 606-8507, Japan
| | - Kenji Yoshida
- Regenerative & Cellular Medicine Kobe Center, Sumitomo Pharma Co., Ltd., Chuo-ku, Kobe 650-0047, Japan
| | - Jun Takahashi
- Department of Clinical Application, Center for iPS Cell Research and Application, Kyoto University, Kyoto 606-8507, Japan.
| |
Collapse
|
3
|
Rasool D, Jahani-Asl A. Master regulators of neurogenesis: the dynamic roles of Ephrin receptors across diverse cellular niches. Transl Psychiatry 2024; 14:462. [PMID: 39505843 PMCID: PMC11541728 DOI: 10.1038/s41398-024-03168-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Revised: 09/20/2024] [Accepted: 10/16/2024] [Indexed: 11/08/2024] Open
Abstract
The ephrin receptors (EphRs) are the largest family of receptor tyrosine kinases (RTKs) that are abundantly expressed in the developing brain and play important roles at different stages of neurogenesis ranging from neural stem cell (NSC) fate specification to neural migration, morphogenesis, and circuit assembly. Defects in EphR signalling have been associated with several pathologies including neurodevelopmental disorders (NDDs), intellectual disability (ID), and neurodegenerative diseases (NDs). Here, we review our current understanding of the complex and dynamic role of EphRs in the brain and discuss how deregulation of these receptors contributes to disease, highlighting their potential as valuable druggable targets.
Collapse
Affiliation(s)
- Dilan Rasool
- Department of Cellular and Molecular Medicine, University of Ottawa, 451 Smyth Road, Ottawa, ON, K1H 8M5, Canada
- University of Ottawa Brain and Mind Research Institute, 451 Smyth Road, Ottawa, ON, K1H 8M5, Canada
- Department of Medicine, Division of Experimental Medicine, McGill University, 1001 Decarie Boulevard, Montreal, QC, H4A 3J1, Canada
- Lady Davis Institute for Medical Research, Jewish General Hospital, 3755 Chemin de la Côte-Sainte-Catherine, Montréal, QC, H3T 1E2, Canada
| | - Arezu Jahani-Asl
- Department of Cellular and Molecular Medicine, University of Ottawa, 451 Smyth Road, Ottawa, ON, K1H 8M5, Canada.
- University of Ottawa Brain and Mind Research Institute, 451 Smyth Road, Ottawa, ON, K1H 8M5, Canada.
- Department of Medicine, Division of Experimental Medicine, McGill University, 1001 Decarie Boulevard, Montreal, QC, H4A 3J1, Canada.
- Lady Davis Institute for Medical Research, Jewish General Hospital, 3755 Chemin de la Côte-Sainte-Catherine, Montréal, QC, H3T 1E2, Canada.
- Gerald Bronfman Department of Oncology, McGill University, 5100 de Maisonneuve Blvd. West, Montréal, QC, H4A 3T2, Canada.
- Regenerative Medicine Program, and Cancer Therapeutics Program, Ottawa Hospital Research Institute, Ottawa, ON, K1H 8L6, Canada.
- Ottawa Institutes of System Biology, University of Ottawa, Health Sciences Campus, 451 Smyth Road, Ottawa, ON, K1H 8M5, Canada.
| |
Collapse
|
4
|
Gupta S, Heinrichs E, Novitch BG, Butler SJ. Investigating the basis of lineage decisions and developmental trajectories in the dorsal spinal cord through pseudotime analyses. Development 2024; 151:dev202209. [PMID: 38804879 PMCID: PMC11166460 DOI: 10.1242/dev.202209] [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: 07/24/2023] [Accepted: 04/18/2024] [Indexed: 05/29/2024]
Abstract
Dorsal interneurons (dIs) in the spinal cord encode the perception of touch, pain, heat, itchiness and proprioception. Previous studies using genetic strategies in animal models have revealed important insights into dI development, but the molecular details of how dIs arise as distinct populations of neurons remain incomplete. We have developed a resource to investigate dI fate specification by combining a single-cell RNA-Seq atlas of mouse embryonic stem cell-derived dIs with pseudotime analyses. To validate this in silico resource as a useful tool, we used it to first identify genes that are candidates for directing the transition states that lead to distinct dI lineage trajectories, and then validated them using in situ hybridization analyses in the developing mouse spinal cord in vivo. We have also identified an endpoint of the dI5 lineage trajectory and found that dIs become more transcriptionally homogeneous during terminal differentiation. This study introduces a valuable tool for further discovery about the timing of gene expression during dI differentiation and demonstrates its utility in clarifying dI lineage relationships.
Collapse
Affiliation(s)
- Sandeep Gupta
- Department of Neurobiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Eric Heinrichs
- Department of Neurobiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Genetics and Genomics Graduate Program, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Bennett G. Novitch
- Department of Neurobiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Intellectual and Developmental Disabilities Research Center, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Samantha J. Butler
- Department of Neurobiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Intellectual and Developmental Disabilities Research Center, University of California, Los Angeles, Los Angeles, CA 90095, USA
| |
Collapse
|
5
|
Gupta S, Heinrichs E, Novitch BG, Butler SJ. Investigating the basis of lineage decisions and developmental trajectories in the dorsal spinal cord through pseudotime analyses. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.07.24.550380. [PMID: 37546781 PMCID: PMC10402035 DOI: 10.1101/2023.07.24.550380] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/08/2023]
Abstract
Dorsal interneurons (dIs) in the spinal cord encode the perception of touch, pain, heat, itch, and proprioception. While previous studies using genetic strategies in animal models have revealed important insights into dI development, the molecular details by which dIs arise as distinct populations of neurons remain incomplete. We have developed a resource to investigate dI fate specification by combining a single-cell RNA-Seq atlas of mouse ESC-derived dIs with pseudotime analyses. To validate this in silico resource as a useful tool, we used it to first identify novel genes that are candidates for directing the transition states that lead to distinct dI lineage trajectories, and then validated them using in situ hybridization analyses in the developing mouse spinal cord in vivo . We have also identified a novel endpoint of the dI5 lineage trajectory and found that dIs become more transcriptionally homogenous during terminal differentiation. Together, this study introduces a valuable tool for further discovery about the timing of gene expression during dI differentiation and demonstrates its utility clarifying dI lineage relationships. Summary statement Pseudotime analyses of embryonic stem cell-derived dorsal spinal interneurons reveals both novel regulators and lineage relationships between different interneuron populations.
Collapse
|
6
|
Brandão-Teles C, Zuccoli GS, de Moraes Vrechi TA, Ramos-da-Silva L, Santos AVS, Crunfli F, Martins-de-Souza D. Induced-pluripotent stem cells and neuroproteomics as tools for studying neurodegeneration. Biochem Soc Trans 2024; 52:163-176. [PMID: 38288874 DOI: 10.1042/bst20230341] [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: 10/16/2023] [Revised: 01/05/2024] [Accepted: 01/08/2024] [Indexed: 02/29/2024]
Abstract
The investigation of neurodegenerative diseases advanced significantly with the advent of cell-reprogramming technology, leading to the creation of new models of human illness. These models, derived from induced pluripotent stem cells (iPSCs), facilitate the study of sporadic as well as hereditary diseases and provide a comprehensive understanding of the molecular mechanisms involved with neurodegeneration. Through proteomics, a quantitative tool capable of identifying thousands of proteins from small sample volumes, researchers have attempted to identify disease mechanisms by detecting differentially expressed proteins and proteoforms in disease models, biofluids, and postmortem brain tissue. The integration of these two technologies allows for the identification of novel pathological targets within the realm of neurodegenerative diseases. Here, we highlight studies from the past 5 years on the contributions of iPSCs within neuroproteomic investigations, which uncover the molecular mechanisms behind these illnesses.
Collapse
Affiliation(s)
- Caroline Brandão-Teles
- Laboratory of Neuroproteomics, Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas, Campinas, Brazil
| | - Giuliana S Zuccoli
- Laboratory of Neuroproteomics, Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas, Campinas, Brazil
| | - Talita Aparecida de Moraes Vrechi
- Laboratory of Neuroproteomics, Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas, Campinas, Brazil
| | - Lívia Ramos-da-Silva
- Laboratory of Neuroproteomics, Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas, Campinas, Brazil
| | - Aline Valéria Sousa Santos
- Laboratory of Neuroproteomics, Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas, Campinas, Brazil
| | - Fernanda Crunfli
- Laboratory of Neuroproteomics, Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas, Campinas, Brazil
| | - Daniel Martins-de-Souza
- Laboratory of Neuroproteomics, Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas, Campinas, Brazil
- Experimental Medicine Research Cluster (EMRC), University of Campinas, Campinas 13083-862, SP, Brazil
- Instituto Nacional de Biomarcadores em Neuropsiquiatria, Conselho Nacional de Desenvolvimento Científico e Tecnológico, São Paulo, Brazil
- INCT in Modelling Human Complex Diseases with 3D Platforms (Model3D)
- D'Or Institute for Research and Education (IDOR), São Paulo, Brazil
| |
Collapse
|
7
|
Klostermeier S, Li A, Hou HX, Green U, Lennerz JK. Exploring the Skin Brain Link: Biomarkers in the Skin with Implications for Aging Research and Alzheimer's Disease Diagnostics. Int J Mol Sci 2023; 24:13309. [PMID: 37686115 PMCID: PMC10487444 DOI: 10.3390/ijms241713309] [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: 07/10/2023] [Revised: 08/08/2023] [Accepted: 08/25/2023] [Indexed: 09/10/2023] Open
Abstract
Neurodegenerative diseases, including Alzheimer's disease (AD), are challenging to diagnose. Currently the field must rely on imperfect diagnostic modalities. A recent study identified differences in several key bio-mechano-physiological parameters of the skin between AD patients and healthy controls. Here, we visually align these differences with the relevant histological, aging, and embryological paradigms to raise awareness for these potential biomarkers. In a study conducted by Wu et al., a series of n = 41 patients (n = 29 with AD and n = 12 healthy controls) were evaluated, demonstrating that AD patients exhibit a less acidic skin pH, increased skin hydration, and reduced skin elasticity compared to healthy controls. We constructed a visual overview and explored the relevant paradigms. We present a visual comparison of these factors, highlighting four paradigms: (1) the findings emphasize a shared ectodermal origin of the brain and the skin; (2) functional systems such as micro-vascularization, innervation, eccrine excretory functions, and the extracellular matrix undergo distinct changes in patients with AD; (3) the human skin mirrors the alterations in brain stiffness observed in aging studies; (4) assessment of physiological features of the skin is cost-effective, accessible, and easily amenable for monitoring and integration with cognitive assessment studies. Understanding the relationship between aging skin and aging brain is an exciting frontier, holding great promise for improved diagnostics. Further prospective and larger-scale investigations are needed to solidify the brain-skin link and determine the extent to which this relationship can be leveraged for diagnostic applications.
Collapse
Affiliation(s)
- Stefanie Klostermeier
- Institute for Physiology and Pathophysiology, University of Erlangen-Nuremberg, 91054 Erlangen, Germany;
| | - Annie Li
- Center for Integrated Diagnostics, Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA; (A.L.); (H.X.H.); (U.G.)
| | - Helen X. Hou
- Center for Integrated Diagnostics, Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA; (A.L.); (H.X.H.); (U.G.)
| | - Ula Green
- Center for Integrated Diagnostics, Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA; (A.L.); (H.X.H.); (U.G.)
| | - Jochen K. Lennerz
- Center for Integrated Diagnostics, Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA; (A.L.); (H.X.H.); (U.G.)
| |
Collapse
|
8
|
Akter M, Ding B. Modeling Movement Disorders via Generation of hiPSC-Derived Motor Neurons. Cells 2022; 11:3796. [PMID: 36497056 PMCID: PMC9737271 DOI: 10.3390/cells11233796] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2022] [Revised: 11/19/2022] [Accepted: 11/24/2022] [Indexed: 11/29/2022] Open
Abstract
Generation of motor neurons (MNs) from human-induced pluripotent stem cells (hiPSCs) overcomes the limited access to human brain tissues and provides an unprecedent approach for modeling MN-related diseases. In this review, we discuss the recent progression in understanding the regulatory mechanisms of MN differentiation and their applications in the generation of MNs from hiPSCs, with a particular focus on two approaches: induction by small molecules and induction by lentiviral delivery of transcription factors. At each induction stage, different culture media and supplements, typical growth conditions and cellular morphology, and specific markers for validation of cell identity and quality control are specifically discussed. Both approaches can generate functional MNs. Currently, the major challenges in modeling neurological diseases using iPSC-derived neurons are: obtaining neurons with high purity and yield; long-term neuron culture to reach full maturation; and how to culture neurons more physiologically to maximize relevance to in vivo conditions.
Collapse
Affiliation(s)
| | - Baojin Ding
- Department of Biochemistry and Molecular Biology, Louisiana State University Health Sciences Center, Shreveport, LA 71130-3932, USA
| |
Collapse
|
9
|
An J, Chen B, Tian D, Guo Y, Yan Y, Yang H. Regulation of Neurogenesis and Neuronal Differentiation by Natural Compounds. Curr Stem Cell Res Ther 2021; 17:756-771. [PMID: 34493197 DOI: 10.2174/1574888x16666210907141447] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Revised: 07/15/2021] [Accepted: 07/28/2021] [Indexed: 11/22/2022]
Abstract
Neuronal damage or degeneration is the main feature of neurological diseases. Regulation of neurogenesis and neuronal differentiation is important in developing therapies to promote neuronal regeneration or synaptic network reconstruction. Neurogenesis is a multistage process in which neurons are generated and integrated into existing neuronal circuits. Neuronal differentiation is extremely complex because it can occur in different cell types and can be caused by a variety of inducers. Recently, natural compounds that induce neurogenesis and neuronal differentiation have attracted extensive attention. In this paper, the potential neural induction effects of medicinal plant-derived natural compounds on neural stem/progenitor cells (NS/PCs), the cultured neuronal cells, and mesenchymal stem cells (MSCs) are reviewed. The natural compounds that are efficacious in inducing neurogenesis and neuronal differentiation include phenolic acids, polyphenols, flavonoids, glucosides, alkaloids, terpenoids, quinones, coumarins, and others. They exert neural induction effects by regulating signal factors and cell-specific genes involved in the process of neurogenesis and neuronal differentiation, including specific proteins (β-tubulin III, MAP-2, tau, nestin, neurofilaments, GFAP, GAP-43, NSE), related genes and proteins (STAT3, Hes1, Mash1, NeuroD1, notch, cyclin D1, SIRT1, reggie-1), transcription factors (CREB, Nkx-2.5, Ngn1), neurotrophins (BDNF, NGF, NT-3) and signaling pathways (JAK/STAT, Wnt/β-catenin, MAPK, PI3K/Akt, GSK-3β/β-catenin, Ca2+/CaMKII/ATF1, Nrf2/HO-1, BMP). The natural compounds with neural induction effects are of great value for neuronal regenerative medicine and provide promising prevention and treatment strategies for neurological diseases.
Collapse
Affiliation(s)
- Jing An
- Translational Medicine Centre, Honghui Hospital, Xi'an Jiaotong University, Xi'an. China
| | - Bo Chen
- Translational Medicine Centre, Honghui Hospital, Xi'an Jiaotong University, Xi'an. China
| | - Ding Tian
- Translational Medicine Centre, Honghui Hospital, Xi'an Jiaotong University, Xi'an. China
| | - Yunshan Guo
- Department of Spine Surgery, Honghui Hospital, Xi'an Jiaotong University, Xi'an. China
| | - Yuzhu Yan
- Clinical Lab, Honghui Hospital, Xi'an Jiaotong University, Xi'an. China
| | - Hao Yang
- Translational Medicine Centre, Honghui Hospital, Xi'an Jiaotong University, Xi'an. China
| |
Collapse
|
10
|
Warnecke A, Giesemann A. Embryology, Malformations, and Rare Diseases of the Cochlea. Laryngorhinootologie 2021; 100:S1-S43. [PMID: 34352899 PMCID: PMC8354575 DOI: 10.1055/a-1349-3824] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Despite the low overall prevalence of individual rare diseases, cochlear
dysfunction leading to hearing loss represents a symptom in a large
proportion. The aim of this work was to provide a clear overview of rare
cochlear diseases, taking into account the embryonic development of the
cochlea and the systematic presentation of the different disorders. Although
rapid biotechnological and bioinformatic advances may facilitate the
diagnosis of a rare disease, an interdisciplinary exchange is often required
to raise the suspicion of a rare disease. It is important to recognize that
the phenotype of rare inner ear diseases can vary greatly not only in
non-syndromic but also in syndromic hearing disorders. Finally, it becomes
clear that the phenotype of the individual rare diseases cannot be
determined exclusively by classical genetics even in monogenetic
disorders.
Collapse
Affiliation(s)
- Athanasia Warnecke
- Klinik für Hals-, Nasen- und Ohrenheilkunde, Medizinische Hochschule Hannover, Carl-Neuberg-Straße 1, 30625 Hannover.,Deutsche Forschungsgemeinschaft Exzellenzcluster"Hearing4all" - EXC 2177/1 - Project ID 390895286
| | - Anja Giesemann
- Institut für Neuroradiologie, Medizinische Hochschule Hannover, Carl-Neuberg-Straße 1, 30625 Hannover
| |
Collapse
|
11
|
Neyrinck K, García-León JA. Single Transcription Factor-Based Differentiation Allowing Fast and Efficient Oligodendrocyte Generation via SOX10 Overexpression. Methods Mol Biol 2021; 2352:149-170. [PMID: 34324186 DOI: 10.1007/978-1-0716-1601-7_11] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Oligodendrocytes are the main glial cell type in the central nervous system supporting the axonal part of neurons via myelin and lactate delivery. Both the conductive myelin formation and the energy support via lactate can be affected in diseases, such as multiple sclerosis and amyotrophic lateral sclerosis, respectively. Therefore, human disease modeling is needed to gain more mechanistic insights to drive drug discovery research. Here, patient-derived induced pluripotent stem cells (iPSCs) serve as a necessary tool providing an infinite cell source for patient-specific disease modeling, which allows investigation of oligodendrocyte involvement in human disease.Small molecule-based differentiation protocols to generate oligodendrocytes from pluripotent stem cells can last more than 90 days. Here, we provide a transcription factor-based, fast and efficient protocol for generating O4+ oligodendrocytes in just 20-24 days. After a neural induction phase of 8-12 days, SOX10 is overexpressed either with the use of lentiviral vectors or via engineered iPSCs, which inducibly overexpress SOX10 after doxycycline addition. Using this last method, a pure O4+ cell population is achieved after keeping the SOX10-overexpressing neural stem cells in culture for an additional 10 days. Furthermore, these O4+ cells can be co-cultured with iPSC-derived cortical neurons in 384-well format, allowing pro-myelinating drug screens. In conclusion, we provide a fast and efficient oligodendrocyte differentiation protocol allowing both in vitro human disease modeling and a high-throughput co-culture system for drug discovery.
Collapse
Affiliation(s)
- Katrien Neyrinck
- Department of Development and Regeneration, Stem Cell Institute, KU Leuven, Leuven, Belgium.
| | - Juan A García-León
- Departamento Biologia Celular, Genetica y Fisiologia, Facultad de Ciencias, Instituto de Investigacion Biomedica de Malaga-IBIMA, Universidad de Malaga, Malaga, Spain
- Centro de Investigacion Biomedica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| |
Collapse
|
12
|
Meisig J, Dreser N, Kapitza M, Henry M, Rotshteyn T, Rahnenführer J, Hengstler J, Sachinidis A, Waldmann T, Leist M, Blüthgen N. Kinetic modeling of stem cell transcriptome dynamics to identify regulatory modules of normal and disturbed neuroectodermal differentiation. Nucleic Acids Res 2020; 48:12577-12592. [PMID: 33245762 PMCID: PMC7736781 DOI: 10.1093/nar/gkaa1089] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Revised: 10/21/2020] [Accepted: 10/27/2020] [Indexed: 12/22/2022] Open
Abstract
Thousands of transcriptome data sets are available, but approaches for their use in dynamic cell response modelling are few, especially for processes affected simultaneously by two orthogonal influencing variables. We approached this problem for neuroepithelial development of human pluripotent stem cells (differentiation variable), in the presence or absence of valproic acid (signaling variable). Using few basic assumptions (sequential differentiation states of cells; discrete on/off states for individual genes in these states), and time-resolved transcriptome data, a comprehensive model of spontaneous and perturbed gene expression dynamics was developed. The model made reliable predictions (average correlation of 0.85 between predicted and subsequently tested expression values). Even regulations predicted to be non-monotonic were successfully validated by PCR in new sets of experiments. Transient patterns of gene regulation were identified from model predictions. They pointed towards activation of Wnt signaling as a candidate pathway leading to a redirection of differentiation away from neuroepithelial cells towards neural crest. Intervention experiments, using a Wnt/beta-catenin antagonist, led to a phenotypic rescue of this disturbed differentiation. Thus, our broadly applicable model allows the analysis of transcriptome changes in complex time/perturbation matrices.
Collapse
Affiliation(s)
- Johannes Meisig
- Institute of Pathology, Charité-Universitätsmedizin, 10117 Berlin, Germany
- IRI Life Sciences, Humboldt-Universität zu Berlin, 10117 Berlin, Germany
| | - Nadine Dreser
- In Vitro Toxicology and Biomedicine, Dept inaugurated by the Doerenkamp-Zbinden Chair foundation, University of Konstanz, 78457 Konstanz, Germany
| | - Marion Kapitza
- In Vitro Toxicology and Biomedicine, Dept inaugurated by the Doerenkamp-Zbinden Chair foundation, University of Konstanz, 78457 Konstanz, Germany
| | - Margit Henry
- Faculty of Medicine, Institute of Neurophysiology, University of Cologne, 50931 Cologne, Germany
- Center for Molecular Medicine Cologne (CMMC), University of Cologne, 50931 Cologne, Germany
| | - Tamara Rotshteyn
- Faculty of Medicine, Institute of Neurophysiology, University of Cologne, 50931 Cologne, Germany
- Center for Molecular Medicine Cologne (CMMC), University of Cologne, 50931 Cologne, Germany
| | - Jörg Rahnenführer
- Department of Statistics, TU Dortmund University, 44221 Dortmund, Germany
| | - Jan G Hengstler
- Leibniz Research Centre for Working Environment and Human Factors (IfADo), TU Dortmund University, 44139 Dortmund, Germany
| | - Agapios Sachinidis
- Faculty of Medicine, Institute of Neurophysiology, University of Cologne, 50931 Cologne, Germany
- Center for Molecular Medicine Cologne (CMMC), University of Cologne, 50931 Cologne, Germany
| | - Tanja Waldmann
- In Vitro Toxicology and Biomedicine, Dept inaugurated by the Doerenkamp-Zbinden Chair foundation, University of Konstanz, 78457 Konstanz, Germany
| | - Marcel Leist
- In Vitro Toxicology and Biomedicine, Dept inaugurated by the Doerenkamp-Zbinden Chair foundation, University of Konstanz, 78457 Konstanz, Germany
| | - Nils Blüthgen
- Institute of Pathology, Charité-Universitätsmedizin, 10117 Berlin, Germany
- IRI Life Sciences, Humboldt-Universität zu Berlin, 10117 Berlin, Germany
| |
Collapse
|
13
|
Identification of Molecular Signatures in Neural Differentiation and Neurological Diseases Using Digital Color-Coded Molecular Barcoding. Stem Cells Int 2020; 2020:8852313. [PMID: 33005195 PMCID: PMC7503121 DOI: 10.1155/2020/8852313] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Revised: 08/26/2020] [Accepted: 08/28/2020] [Indexed: 12/04/2022] Open
Abstract
Human pluripotent stem cells (PSCs), including embryonic stem cells and induced pluripotent stem cells, represent powerful tools for disease modeling and for therapeutic applications. PSCs are particularly useful for the study of development and diseases of the nervous system. However, generating in vitro models that recapitulate the architecture and the full variety of subtypes of cells that make the complexity of our brain remains a challenge. In order to fully exploit the potential of PSCs, advanced methods that facilitate the identification of molecular signatures in neural differentiation and neurological diseases are highly demanded. Here, we review the literature on the development and application of digital color-coded molecular barcoding as a potential tool for standardizing PSC research and applications in neuroscience. We will also describe relevant examples of the use of this technique for the characterization of the heterogeneous composition of the brain tumor glioblastoma multiforme.
Collapse
|
14
|
Kim YK, Lee H, Ismail T, Kim Y, Lee HS. Dach1 regulates neural crest migration during embryonic development. Biochem Biophys Res Commun 2020; 527:896-901. [PMID: 32430182 DOI: 10.1016/j.bbrc.2020.05.009] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Accepted: 05/02/2020] [Indexed: 02/06/2023]
Abstract
Dachshund 1(Dach1) is a key component of the retinal determination gene network that plays significant roles in cell fate regulation. The vertebrate homolog of Drosophila dachshund has gained considerable importance as an essential regulator of development, but its functions during embryonic development remain elusive. We investigated the functional significance of dach1 during Xenopus embryogenesis using loss-of-function studies. Reverse transcription-polymerase chain reaction demonstrated the maternal nature of dach1, showing enhanced expression at the neurula stage of development, and morpholino oligonucleotide injection of dach1 induced phenotypic anomalies of microcephaly and reduced body length. Animal cap assays followed by whole-mount in-situ hybridization indicated the perturbed expression of neural and neural crest (NC) markers. Our data suggest the prerequisite functions of dach1 in NC migration during Xenopus embryogenesis. However, the developmental pathways regulated by dach1 during embryogenesis require further elucidation.
Collapse
Affiliation(s)
- Yoo-Kyung Kim
- KNU-Center for Nonlinear Dynamics, CMRI, School of Life Sciences, BK21 Plus KNU Creative BioResearch Group, College of Natural Sciences, Kyungpook National University, Daegu, 41566, South Korea
| | - Hongchan Lee
- KNU-Center for Nonlinear Dynamics, CMRI, School of Life Sciences, BK21 Plus KNU Creative BioResearch Group, College of Natural Sciences, Kyungpook National University, Daegu, 41566, South Korea
| | - Tayaba Ismail
- KNU-Center for Nonlinear Dynamics, CMRI, School of Life Sciences, BK21 Plus KNU Creative BioResearch Group, College of Natural Sciences, Kyungpook National University, Daegu, 41566, South Korea
| | - Youni Kim
- KNU-Center for Nonlinear Dynamics, CMRI, School of Life Sciences, BK21 Plus KNU Creative BioResearch Group, College of Natural Sciences, Kyungpook National University, Daegu, 41566, South Korea
| | - Hyun-Shik Lee
- KNU-Center for Nonlinear Dynamics, CMRI, School of Life Sciences, BK21 Plus KNU Creative BioResearch Group, College of Natural Sciences, Kyungpook National University, Daegu, 41566, South Korea.
| |
Collapse
|
15
|
Wegner SH, Park JJ, Workman T, Hermsen SAB, Wallace J, Stanaway IB, Kim HY, Griffith WC, Hong S, Faustman EM. Anchoring a dynamic in vitro model of human neuronal differentiation to key processes of early brain development in vivo. Reprod Toxicol 2020; 91:116-130. [PMID: 31740287 PMCID: PMC6980388 DOI: 10.1016/j.reprotox.2019.09.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Revised: 09/25/2019] [Accepted: 09/27/2019] [Indexed: 01/04/2023]
Abstract
We characterize temporal pathway dynamics of differentiation in an in vitro neurotoxicity model with the aim of informing design and interpretation of toxicological assays. Human neural progenitor cells (hNPCs) were cultured in differentiation conditions up to 21 days. Genes significantly changed through time were identified and grouped according to temporal dynamics. Quantitative pathway analysis identified gene ontology (GO) terms enriched among significantly changed genes and provided a temporal roadmap of pathway trends in vitro. Gene expression in hNPCs was compared with publicly available gene expression data from developing human brain tissue in vivo. Quantitative pathway analysis of significantly changed genes and targeted analysis of specific pathways of interest identified concordance between in vivo and in vitro expression associated with proliferation, migration, differentiation, synapse formation, and neurotransmission. Our analysis anchors gene expression patterns in vitro to sensitive windows of in vivo development, helping to define appropriate applications of the model.
Collapse
Affiliation(s)
- Susanna H Wegner
- Institute for Risk Analysis and Risk Communication, Department of Environmental and Occupational Health Sciences, School of Public Health, University of Washington, Seattle, WA, United States
| | - Julie Juyoung Park
- Institute for Risk Analysis and Risk Communication, Department of Environmental and Occupational Health Sciences, School of Public Health, University of Washington, Seattle, WA, United States
| | - Tomomi Workman
- Institute for Risk Analysis and Risk Communication, Department of Environmental and Occupational Health Sciences, School of Public Health, University of Washington, Seattle, WA, United States
| | - Sanne A B Hermsen
- Institute for Risk Analysis and Risk Communication, Department of Environmental and Occupational Health Sciences, School of Public Health, University of Washington, Seattle, WA, United States
| | - Jim Wallace
- Institute for Risk Analysis and Risk Communication, Department of Environmental and Occupational Health Sciences, School of Public Health, University of Washington, Seattle, WA, United States
| | - Ian B Stanaway
- Institute for Risk Analysis and Risk Communication, Department of Environmental and Occupational Health Sciences, School of Public Health, University of Washington, Seattle, WA, United States
| | - Hee Yeon Kim
- Institute for Risk Analysis and Risk Communication, Department of Environmental and Occupational Health Sciences, School of Public Health, University of Washington, Seattle, WA, United States
| | - William C Griffith
- Institute for Risk Analysis and Risk Communication, Department of Environmental and Occupational Health Sciences, School of Public Health, University of Washington, Seattle, WA, United States
| | - Sungwoo Hong
- Institute for Risk Analysis and Risk Communication, Department of Environmental and Occupational Health Sciences, School of Public Health, University of Washington, Seattle, WA, United States
| | - Elaine M Faustman
- Institute for Risk Analysis and Risk Communication, Department of Environmental and Occupational Health Sciences, School of Public Health, University of Washington, Seattle, WA, United States.
| |
Collapse
|
16
|
Linaro D, Vermaercke B, Iwata R, Ramaswamy A, Libé-Philippot B, Boubakar L, Davis BA, Wierda K, Davie K, Poovathingal S, Penttila PA, Bilheu A, De Bruyne L, Gall D, Conzelmann KK, Bonin V, Vanderhaeghen P. Xenotransplanted Human Cortical Neurons Reveal Species-Specific Development and Functional Integration into Mouse Visual Circuits. Neuron 2019; 104:972-986.e6. [PMID: 31761708 PMCID: PMC6899440 DOI: 10.1016/j.neuron.2019.10.002] [Citation(s) in RCA: 126] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2019] [Revised: 08/12/2019] [Accepted: 09/30/2019] [Indexed: 12/31/2022]
Abstract
How neural circuits develop in the human brain has remained almost impossible to study at the neuronal level. Here, we investigate human cortical neuron development, plasticity, and function using a mouse/human chimera model in which xenotransplanted human cortical pyramidal neurons integrate as single cells into the mouse cortex. Combined neuronal tracing, electrophysiology, and in vivo structural and functional imaging of the transplanted cells reveal a coordinated developmental roadmap recapitulating key milestones of human cortical neuron development. The human neurons display a prolonged developmental timeline, indicating the neuron-intrinsic retention of juvenile properties as an important component of human brain neoteny. Following maturation, human neurons in the visual cortex display tuned, decorrelated responses to visual stimuli, like mouse neurons, demonstrating their capacity for physiological synaptic integration in host cortical circuits. These findings provide new insights into human neuronal development and open novel avenues for the study of human neuronal function and disease. Video Abstract
Cell-intrinsic mechanisms of human neoteny in mouse-human chimeric cerebral cortex Human neurons show prolonged maturation and single-cell integration in mouse cortex Stable dendritic spines and long-term synaptic plasticity in xenotransplanted neurons Human neurons show decorrelated activity and tuned responses to visual stimuli
Collapse
Affiliation(s)
- Daniele Linaro
- VIB-KU Leuven Center for Brain & Disease Research, 3000 Leuven, Belgium; Department of Neurosciences and Leuven Brain Institute, KU Leuven, 3000 Leuven, Belgium; Institut de Recherches en Biologie Humaine et Moléculaire (IRIBHM) and ULB Neuroscience Institute (UNI), Université Libre de Bruxelles (ULB), 1070 Brussels, Belgium
| | - Ben Vermaercke
- VIB-KU Leuven Center for Brain & Disease Research, 3000 Leuven, Belgium; Department of Neurosciences and Leuven Brain Institute, KU Leuven, 3000 Leuven, Belgium; Neuro-Electronics Research Flanders, Kapeldreef 75, 3001 Leuven, Belgium
| | - Ryohei Iwata
- VIB-KU Leuven Center for Brain & Disease Research, 3000 Leuven, Belgium; Department of Neurosciences and Leuven Brain Institute, KU Leuven, 3000 Leuven, Belgium; Institut de Recherches en Biologie Humaine et Moléculaire (IRIBHM) and ULB Neuroscience Institute (UNI), Université Libre de Bruxelles (ULB), 1070 Brussels, Belgium
| | - Arjun Ramaswamy
- Department of Neurosciences and Leuven Brain Institute, KU Leuven, 3000 Leuven, Belgium; Neuro-Electronics Research Flanders, Kapeldreef 75, 3001 Leuven, Belgium; imec, 3001 Leuven, Belgium
| | - Baptiste Libé-Philippot
- VIB-KU Leuven Center for Brain & Disease Research, 3000 Leuven, Belgium; Department of Neurosciences and Leuven Brain Institute, KU Leuven, 3000 Leuven, Belgium; Institut de Recherches en Biologie Humaine et Moléculaire (IRIBHM) and ULB Neuroscience Institute (UNI), Université Libre de Bruxelles (ULB), 1070 Brussels, Belgium
| | - Leila Boubakar
- VIB-KU Leuven Center for Brain & Disease Research, 3000 Leuven, Belgium; Department of Neurosciences and Leuven Brain Institute, KU Leuven, 3000 Leuven, Belgium; Institut de Recherches en Biologie Humaine et Moléculaire (IRIBHM) and ULB Neuroscience Institute (UNI), Université Libre de Bruxelles (ULB), 1070 Brussels, Belgium
| | - Brittany A Davis
- Institut de Recherches en Biologie Humaine et Moléculaire (IRIBHM) and ULB Neuroscience Institute (UNI), Université Libre de Bruxelles (ULB), 1070 Brussels, Belgium
| | - Keimpe Wierda
- VIB-KU Leuven Center for Brain & Disease Research, 3000 Leuven, Belgium
| | - Kristofer Davie
- VIB-KU Leuven Center for Brain & Disease Research, 3000 Leuven, Belgium
| | | | | | - Angéline Bilheu
- Institut de Recherches en Biologie Humaine et Moléculaire (IRIBHM) and ULB Neuroscience Institute (UNI), Université Libre de Bruxelles (ULB), 1070 Brussels, Belgium
| | - Lore De Bruyne
- VIB-KU Leuven Center for Brain & Disease Research, 3000 Leuven, Belgium; Department of Neurosciences and Leuven Brain Institute, KU Leuven, 3000 Leuven, Belgium
| | - David Gall
- Institut de Recherches en Biologie Humaine et Moléculaire (IRIBHM) and ULB Neuroscience Institute (UNI), Université Libre de Bruxelles (ULB), 1070 Brussels, Belgium; Laboratoire de Physiologie et Pharmacologie and ULB Neuroscience Institute (UNI), Université Libre de Bruxelles (ULB), 1070 Brussels, Belgium
| | - Karl-Klaus Conzelmann
- Max von Pettenkofer-Institute and Gene Center, Ludwig-Maximilians-Universität Munchen, 81377 Munich, Germany
| | - Vincent Bonin
- Neuro-Electronics Research Flanders, Kapeldreef 75, 3001 Leuven, Belgium; imec, 3001 Leuven, Belgium; Department of Biology and Leuven Brain Institute, KU Leuven, 3000 Leuven, Belgium; VIB, 3000 Leuven, Belgium.
| | - Pierre Vanderhaeghen
- VIB-KU Leuven Center for Brain & Disease Research, 3000 Leuven, Belgium; Department of Neurosciences and Leuven Brain Institute, KU Leuven, 3000 Leuven, Belgium; Institut de Recherches en Biologie Humaine et Moléculaire (IRIBHM) and ULB Neuroscience Institute (UNI), Université Libre de Bruxelles (ULB), 1070 Brussels, Belgium; VIB, 3000 Leuven, Belgium; Welbio, Université Libre de Bruxelles (ULB), 1070 Brussels, Belgium.
| |
Collapse
|
17
|
Dabrowski A, Robinson TJ, Felling RJ. Promoting Brain Repair and Regeneration After Stroke: a Plea for Cell-Based Therapies. Curr Neurol Neurosci Rep 2019; 19:5. [PMID: 30712068 DOI: 10.1007/s11910-019-0920-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
PURPOSE OF REVIEW After decades of hype, cell-based therapies are emerging into the clinical arena for the purposes of promoting recovery after stroke. In this review, we discuss the most recent science behind the role of cell-based therapies in ischemic stroke and the efforts to translate these therapies into human clinical trials. RECENT FINDINGS Preclinical data support numerous beneficial effects of cell-based therapies in both small and large animal models of ischemic stroke. These benefits are driven by multifaceted mechanisms promoting brain repair through immunomodulation, trophic support, circuit reorganization, and cell replacement. Cell-based therapies offer tremendous potential for improving outcomes after stroke through multimodal support of brain repair. Based on recent clinical trials, cell-based therapies appear both feasible and safe in all phases of stroke. Ongoing translational research and clinical trials will further refine these therapies and have the potential to transform the approach to stroke recovery and rehabilitation.
Collapse
Affiliation(s)
- Ania Dabrowski
- Department of Neurology, Johns Hopkins School of Medicine, 200 N. Wolfe Street, Suite 2158, Baltimore, MD, 21287, USA
| | - Thomas J Robinson
- Department of Neurology, Johns Hopkins School of Medicine, 200 N. Wolfe Street, Suite 2158, Baltimore, MD, 21287, USA
| | - Ryan J Felling
- Department of Neurology, Johns Hopkins School of Medicine, 200 N. Wolfe Street, Suite 2158, Baltimore, MD, 21287, USA.
| |
Collapse
|
18
|
Skin Stem Cells, Their Niche and Tissue Engineering Approach for Skin Regeneration. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1212:107-126. [PMID: 31065940 DOI: 10.1007/5584_2019_380] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Skin is the main organ that covers the human body and acts as a protective barrier between the human body and the environment. Skin tissue as a stem cell source can be used for transplantation in therapeutic application in terms of its properties such as abundant, easy to access, high plasticity and high ability to regenerate. The immunological profile of these cells makes it a suitable resource for autologous and allogeneic applications. The lack of major histo-compatibility complex 1 is also advantageous in its use. Epidermal stem cells are the main stem cells in the skin and are suitable cells in tissue engineering studies for their important role in wound repair. In the last 30 years, many studies have been conducted to develop substitutions that mimic human skin. Stem cell-based skin substitutions have been developed to be used in clinical applications, to support the healing of acute and chronic wounds and as test systems for dermatological and pharmacological applications. In this chapter, tissue specific properties of epidermal stem cells, composition of their niche, regenerative approaches and repair of tissue degeneration have been examined.
Collapse
|
19
|
Kishi Y, Gotoh Y. Regulation of Chromatin Structure During Neural Development. Front Neurosci 2018; 12:874. [PMID: 30618540 PMCID: PMC6297780 DOI: 10.3389/fnins.2018.00874] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2018] [Accepted: 11/09/2018] [Indexed: 11/13/2022] Open
Abstract
The regulation of genome architecture is a key determinant of gene transcription patterns and neural development. Advances in methodologies based on chromatin conformation capture (3C) have shed light on the genome-wide organization of chromatin in developmental processes. Here, we review recent discoveries regarding the regulation of three-dimensional (3D) chromatin conformation, including promoter-enhancer looping, and the dynamics of large chromatin domains such as topologically associated domains (TADs) and A/B compartments. We conclude with perspectives on how these conformational changes govern neural development and may go awry in disease states.
Collapse
Affiliation(s)
- Yusuke Kishi
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| | - Yukiko Gotoh
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
- International Research Center for Neurointelligence (WPI-IRCN), The University of Tokyo, Tokyo, Japan
| |
Collapse
|
20
|
Kim JY, Lee JS, Hwang HS, Lee DR, Park CY, Jung SJ, You YR, Kim DS, Kim DW. Wnt signal activation induces midbrain specification through direct binding of the beta-catenin/TCF4 complex to the EN1 promoter in human pluripotent stem cells. Exp Mol Med 2018; 50:1-13. [PMID: 29650976 PMCID: PMC5938028 DOI: 10.1038/s12276-018-0044-y] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2017] [Revised: 11/30/2017] [Accepted: 12/19/2017] [Indexed: 12/12/2022] Open
Abstract
The canonical Wnt signal pathway plays a pivotal role in anteroposterior patterning and midbrain specification during early neurogenesis. Activating Wnt signal has been a strategy for differentiating human pluripotent stem cells (PSCs) into midbrain dopaminergic (DA) neurons; however, the underlying molecular mechanism(s) of how the Wnt signal drives posterior fate remained unclear. In this study, we found that activating the canonical Wnt signal significantly upregulated the expression of EN1, a midbrain-specific marker, in a fibroblast growth factor signal-dependent manner in human PSC-derived neural precursor cells (NPCs). The EN1 promoter region contains a putative TCF4-binding site that directly interacts with the β-catenin/TCF complex upon Wnt signal activation. Once differentiated, NPCs treated with a Wnt signal agonist gave rise to functional midbrain neurons including glutamatergic, GABAergic, and DA neurons. Our results provide a potential molecular mechanism that underlies midbrain specification of human PSC-derived NPCs by Wnt activation, as well as a differentiation paradigm for generating human midbrain neurons that may serve as a cellular platform for studying the ontogenesis of midbrain neurons and neurological diseases relevant to the midbrain.
Collapse
Affiliation(s)
- Ji Young Kim
- Department of Physiology, Yonsei University College of Medicine, 50-1 Yonsei-ro Seodaemun-gu, Seoul, 03722, Korea.,Brain Korea 21 PLUS Program for Medical Science, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Korea
| | - Jae Souk Lee
- Department of Physiology, Yonsei University College of Medicine, 50-1 Yonsei-ro Seodaemun-gu, Seoul, 03722, Korea.,Brain Korea 21 PLUS Program for Medical Science, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Korea
| | - Hyun Sub Hwang
- Department of Physiology, Yonsei University College of Medicine, 50-1 Yonsei-ro Seodaemun-gu, Seoul, 03722, Korea
| | - Dongjin R Lee
- Department of Physiology, Yonsei University College of Medicine, 50-1 Yonsei-ro Seodaemun-gu, Seoul, 03722, Korea
| | - Chul-Yong Park
- Department of Physiology, Yonsei University College of Medicine, 50-1 Yonsei-ro Seodaemun-gu, Seoul, 03722, Korea
| | - Sung Jun Jung
- Department of Physiology, College of Medicine, Hanyang University, 222 Wangsimni-ro, Seoul, 04763, Korea
| | - Young Rang You
- Department of Biotechnology, Brain Korea 21 PLUS program for Biotechnology, College of Life Science & Biotechnology, Korea University, 145 Anam-ro, Seongbuk-gu, Seoul, 02841, Korea
| | - Dae-Sung Kim
- Department of Biotechnology, Brain Korea 21 PLUS program for Biotechnology, College of Life Science & Biotechnology, Korea University, 145 Anam-ro, Seongbuk-gu, Seoul, 02841, Korea.
| | - Dong-Wook Kim
- Department of Physiology, Yonsei University College of Medicine, 50-1 Yonsei-ro Seodaemun-gu, Seoul, 03722, Korea. .,Brain Korea 21 PLUS Program for Medical Science, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Korea.
| |
Collapse
|
21
|
Characterization of 3D embryonic C57BL/6 and A/J mouse midbrain micromass in vitro culture systems for developmental neurotoxicity testing. Toxicol In Vitro 2018; 48:33-44. [DOI: 10.1016/j.tiv.2017.12.009] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2017] [Accepted: 12/13/2017] [Indexed: 01/19/2023]
|
22
|
|
23
|
Alsanie WF, Niclis JC, Hunt CP, De Luzy IR, Penna V, Bye CR, Pouton CW, Haynes J, Firas J, Thompson LH, Parish CL. Specification of murine ground state pluripotent stem cells to regional neuronal populations. Sci Rep 2017; 7:16001. [PMID: 29167563 PMCID: PMC5700195 DOI: 10.1038/s41598-017-16248-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2017] [Accepted: 11/08/2017] [Indexed: 11/20/2022] Open
Abstract
Pluripotent stem cells (PSCs) are a valuable tool for interrogating development, disease modelling, drug discovery and transplantation. Despite the burgeoned capability to fate restrict human PSCs to specific neural lineages, comparative protocols for mouse PSCs have not similarly advanced. Mouse protocols fail to recapitulate neural development, consequently yielding highly heterogeneous populations, yet mouse PSCs remain a valuable scientific tool as differentiation is rapid, cost effective and an extensive repertoire of transgenic lines provides an invaluable resource for understanding biology. Here we developed protocols for neural fate restriction of mouse PSCs, using knowledge of embryonic development and recent progress with human equivalents. These methodologies rely upon naïve ground-state PSCs temporarily transitioning through LIF-responsive stage prior to neural induction and rapid exposure to regional morphogens. Neural subtypes generated included those of the dorsal forebrain, ventral forebrain, ventral midbrain and hindbrain. This rapid specification, without feeder layers or embryoid-body formation, resulted in high proportions of correctly specified progenitors and neurons with robust reproducibility. These generated neural progenitors/neurons will provide a valuable resource to further understand development, as well disorders affecting specific neuronal subpopulations.
Collapse
Affiliation(s)
- Walaa F Alsanie
- The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Melbourne, Australia.,The Department of Medical Laboratories, The Faculty of Applied Medical Sciences, Taif University, Taif, Saudi Arabia
| | - Jonathan C Niclis
- The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Melbourne, Australia
| | - Cameron P Hunt
- The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Melbourne, Australia.,Monash Institute of Pharmaceutical Sciences, Monash University, Melbourne, Australia
| | - Isabelle R De Luzy
- The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Melbourne, Australia
| | - Vanessa Penna
- The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Melbourne, Australia
| | - Christopher R Bye
- The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Melbourne, Australia
| | - Colin W Pouton
- Monash Institute of Pharmaceutical Sciences, Monash University, Melbourne, Australia
| | - John Haynes
- Monash Institute of Pharmaceutical Sciences, Monash University, Melbourne, Australia
| | - Jaber Firas
- The Australian Regenerative Medicine Institute, Monash University, Melbourne, Australia
| | - Lachlan H Thompson
- The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Melbourne, Australia
| | - Clare L Parish
- The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Melbourne, Australia.
| |
Collapse
|
24
|
Cruz-Molina S, Respuela P, Tebartz C, Kolovos P, Nikolic M, Fueyo R, van Ijcken WF, Grosveld F, Frommolt P, Bazzi H, Rada-Iglesias A. PRC2 Facilitates the Regulatory Topology Required for Poised Enhancer Function during Pluripotent Stem Cell Differentiation. Cell Stem Cell 2017; 20:689-705.e9. [DOI: 10.1016/j.stem.2017.02.004] [Citation(s) in RCA: 151] [Impact Index Per Article: 18.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2016] [Revised: 12/19/2016] [Accepted: 02/07/2017] [Indexed: 01/28/2023]
|
25
|
Sherman JH, Karpinski BA, Fralish MS, Cappuzzo JM, Dhindsa DS, Thal AG, Moody SA, LaMantia AS, Maynard TM. Foxd4 is essential for establishing neural cell fate and for neuronal differentiation. Genesis 2017; 55. [PMID: 28316121 DOI: 10.1002/dvg.23031] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2017] [Revised: 03/10/2017] [Accepted: 03/13/2017] [Indexed: 01/21/2023]
Abstract
Many molecular factors required for later stages of neuronal differentiation have been identified; however, much less is known about the early events that regulate the initial establishment of the neuroectoderm. We have used an in vitro embryonic stem cell (ESC) differentiation model to investigate early events of neuronal differentiation and to define the role of mouse Foxd4, an ortholog of a forkhead-family transcription factor central to Xenopus neural plate/neuroectodermal precursor development. We found that Foxd4 is a necessary regulator of the transition from pluripotent ESC to neuroectodermal stem cell, and its expression is necessary for neuronal differentiation. Mouse Foxd4 expression is not only limited to the neural plate but it is also expressed and apparently functions to regulate neurogenesis in the olfactory placode. These in vitro results suggest that mouse Foxd4 has a similar function to its Xenopus ortholog; this was confirmed by successfully substituting murine Foxd4 for its amphibian counterpart in overexpression experiments. Thus, Foxd4 appears to regulate the initial steps in establishing neuroectodermal precursors during initial development of the nervous system.
Collapse
Affiliation(s)
- Jonathan H Sherman
- Department of Neurological Surgery, George Washington University Hospital, Washington, District of Columbia.,Institute for Neuroscience, George Washington University, Washington, District of Columbia
| | - Beverly A Karpinski
- Institute for Neuroscience, George Washington University, Washington, District of Columbia.,Department of Pharmacology and Physiology, George Washington University SMHS, Washington, District of Columbia
| | - Matthew S Fralish
- Institute for Neuroscience, George Washington University, Washington, District of Columbia.,Department of Pharmacology and Physiology, George Washington University SMHS, Washington, District of Columbia
| | | | | | - Arielle G Thal
- George Washington University SMHS, Washington, District of Columbia
| | - Sally A Moody
- Institute for Neuroscience, George Washington University, Washington, District of Columbia.,Department of Anatomy and Regenerative Biology, George Washington University SMHS, Washington, District of Columbia
| | - Anthony S LaMantia
- Institute for Neuroscience, George Washington University, Washington, District of Columbia.,Department of Pharmacology and Physiology, George Washington University SMHS, Washington, District of Columbia
| | - Thomas M Maynard
- Institute for Neuroscience, George Washington University, Washington, District of Columbia.,Department of Pharmacology and Physiology, George Washington University SMHS, Washington, District of Columbia
| |
Collapse
|
26
|
Sankar S, Yellajoshyula D, Zhang B, Teets B, Rockweiler N, Kroll KL. Gene regulatory networks in neural cell fate acquisition from genome-wide chromatin association of Geminin and Zic1. Sci Rep 2016; 6:37412. [PMID: 27881878 PMCID: PMC5121602 DOI: 10.1038/srep37412] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2016] [Accepted: 10/28/2016] [Indexed: 12/30/2022] Open
Abstract
Neural cell fate acquisition is mediated by transcription factors expressed in nascent neuroectoderm, including Geminin and members of the Zic transcription factor family. However, regulatory networks through which this occurs are not well defined. Here, we identified Geminin-associated chromatin locations in embryonic stem cells and Geminin- and Zic1-associated locations during neural fate acquisition at a genome-wide level. We determined how Geminin deficiency affected histone acetylation at gene promoters during this process. We integrated these data to demonstrate that Geminin associates with and promotes histone acetylation at neurodevelopmental genes, while Geminin and Zic1 bind a shared gene subset. Geminin- and Zic1-associated genes exhibit embryonic nervous system-enriched expression and encode other regulators of neural development. Both Geminin and Zic1-associated peaks are enriched for Zic1 consensus binding motifs, while Zic1-bound peaks are also enriched for Sox3 motifs, suggesting co-regulatory potential. Accordingly, we found that Geminin and Zic1 could cooperatively activate the expression of several shared targets encoding transcription factors that control neurogenesis, neural plate patterning, and neuronal differentiation. We used these data to construct gene regulatory networks underlying neural fate acquisition. Establishment of this molecular program in nascent neuroectoderm directly links early neural cell fate acquisition with regulatory control of later neurodevelopment.
Collapse
Affiliation(s)
- Savita Sankar
- Department of Developmental Biology, Washington University School of Medicine, 660 S. Euclid Avenue, Saint Louis, MO 63110, USA
| | - Dhananjay Yellajoshyula
- Department of Developmental Biology, Washington University School of Medicine, 660 S. Euclid Avenue, Saint Louis, MO 63110, USA
| | - Bo Zhang
- Department of Developmental Biology, Washington University School of Medicine, 660 S. Euclid Avenue, Saint Louis, MO 63110, USA
| | - Bryan Teets
- Department of Developmental Biology, Washington University School of Medicine, 660 S. Euclid Avenue, Saint Louis, MO 63110, USA
| | - Nicole Rockweiler
- Department of Genetics, Washington University School of Medicine, 660 S. Euclid Avenue, Saint Louis, MO 63110, USA
| | - Kristen L Kroll
- Department of Developmental Biology, Washington University School of Medicine, 660 S. Euclid Avenue, Saint Louis, MO 63110, USA
| |
Collapse
|
27
|
Shroff G. Morphogenesis of human embryonic stem cells into mature neurons under in vitro culture conditions. World J Exp Med 2016; 6:72-79. [PMID: 27909687 PMCID: PMC5114434 DOI: 10.5493/wjem.v6.i4.72] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2016] [Revised: 05/25/2016] [Accepted: 07/18/2016] [Indexed: 02/06/2023] Open
Abstract
AIM To describe the morphogenesis of different neuronal cells from the human embryonic stem cell (hESC) line, SCT-N, under in vitro culture conditions.
METHODS The directed neuronal cell line was produced from a single, spare, pre-implantation stage fertilized ovum that was obtained during a natural in vitro fertilization process. The hESCs were cultured and maintained as per our proprietary in-house technology in a Good Manufacturing Practice, Good Laboratory Practice and Good Tissue Practice compliant laboratory. The cell line was derived and incubated in aerobic conditions. The cells were examined daily under a phase contrast microscope for their growth and differentiation.
RESULTS Different neural progenitor cells (NPCs) and differentiating neurons were observed under the culture conditions. Multipotent NPCs differentiated into all three types of nervous system cells, i.e., neurons, oligodendrocytes and astrocytes. Small projections resembling neurites or dendrites, and protrusion coming out of the cells, were observed. Differentiating cells were observed at day 18 to 20. The differentiating neurons, neuronal bodies, axons, and neuronal tissue were observed on day 21 and day 30 of the culture. On day 25 and day 30, prominent neurons, axons and neuronal tissue were observed under phase contrast microscopy. 4’, 6-diamidino-2-phenylindole staining also indicated the pattern of differentiating neurons, axonal structure and neuronal tissue.
CONCLUSION This study describes the generation of different neuronal cells from an hESC line derived from biopsy of blastomeres at the two-cell cleavage stage from a discarded embryo.
Collapse
|
28
|
Modeling ALS with motor neurons derived from human induced pluripotent stem cells. Nat Neurosci 2016; 19:542-53. [PMID: 27021939 DOI: 10.1038/nn.4273] [Citation(s) in RCA: 200] [Impact Index Per Article: 22.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2015] [Accepted: 02/22/2016] [Indexed: 02/08/2023]
Abstract
Directing the differentiation of induced pluripotent stem cells into motor neurons has allowed investigators to develop new models of amyotrophic lateral sclerosis (ALS). However, techniques vary between laboratories and the cells do not appear to mature into fully functional adult motor neurons. Here we discuss common developmental principles of both lower and upper motor neuron development that have led to specific derivation techniques. We then suggest how these motor neurons may be matured further either through direct expression or administration of specific factors or coculture approaches with other tissues. Ultimately, through a greater understanding of motor neuron biology, it will be possible to establish more reliable models of ALS. These in turn will have a greater chance of validating new drugs that may be effective for the disease.
Collapse
|
29
|
Insights into the Biology and Therapeutic Applications of Neural Stem Cells. Stem Cells Int 2016; 2016:9745315. [PMID: 27069486 PMCID: PMC4812498 DOI: 10.1155/2016/9745315] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2015] [Accepted: 02/08/2016] [Indexed: 12/27/2022] Open
Abstract
The cerebral cortex is essential for our higher cognitive functions and emotional reasoning. Arguably, this brain structure is the distinguishing feature of our species, and yet our remarkable cognitive capacity has seemingly come at a cost to the regenerative capacity of the human brain. Indeed, the capacity for regeneration and neurogenesis of the brains of vertebrates has declined over the course of evolution, from fish to rodents to primates. Nevertheless, recent evidence supporting the existence of neural stem cells (NSCs) in the adult human brain raises new questions about the biological significance of adult neurogenesis in relation to ageing and the possibility that such endogenous sources of NSCs might provide therapeutic options for the treatment of brain injury and disease. Here, we highlight recent insights and perspectives on NSCs within both the developing and adult cerebral cortex. Our review of NSCs during development focuses upon the diversity and therapeutic potential of these cells for use in cellular transplantation and in the modeling of neurodevelopmental disorders. Finally, we describe the cellular and molecular characteristics of NSCs within the adult brain and strategies to harness the therapeutic potential of these cell populations in the treatment of brain injury and disease.
Collapse
|
30
|
Hao B, Webb SE, Miller AL, Yue J. The role of Ca(2+) signaling on the self-renewal and neural differentiation of embryonic stem cells (ESCs). Cell Calcium 2016; 59:67-74. [PMID: 26973143 DOI: 10.1016/j.ceca.2016.01.004] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2015] [Revised: 01/05/2016] [Accepted: 01/19/2016] [Indexed: 12/12/2022]
Abstract
Embryonic stem cells (ESCs) are promising resources for both scientific research and clinical regenerative medicine. With regards to the latter, ESCs are especially useful for treating several neurodegenerative disorders. Two significant characteristics of ESCs, which make them so valuable, are their capacity for self-renewal and their pluripotency, both of which are regulated by the integration of various signaling pathways. Intracellular Ca(2+) signaling is involved in several of these pathways. It is known to be precisely controlled by different Ca(2+) channels and pumps, which play an important role in a variety of cellular activities, including proliferation, differentiation and apoptosis. Here, we provide a review of the recent work conducted to investigate the function of Ca(2+) signaling in the self-renewal and the neural differentiation of ESCs. Specifically, we describe the role of intracellular Ca(2+) mobilization mediated by RyRs (ryanodine receptors); by cADPR (cyclic adenosine 5'-diphosphate ribose) and CD38 (cluster of differentiation 38/cADPR hydrolase); and by NAADP (nicotinic acid adenine dinucleotide phosphate) and TPC2 (two pore channel 2). We also discuss the Ca(2+) influx mediated by SOCs (store-operated Ca(2+) channels), TRPCs (transient receptor potential cation channels) and LTCC (L-type Ca(2+) channels) in the pluripotent ESCs as well as in neural differentiation of ESCs. Moreover, we describe the integration of Ca(2+) signaling in the other signaling pathways that are known to regulate the fate of ESCs.
Collapse
Affiliation(s)
- Baixia Hao
- Division of Life Science and State Key Laboratory of Molecular Neuroscience, HKUST, Clear Water Bay, Hong Kong, China
| | - Sarah E Webb
- Division of Life Science and State Key Laboratory of Molecular Neuroscience, HKUST, Clear Water Bay, Hong Kong, China
| | - Andrew L Miller
- Division of Life Science and State Key Laboratory of Molecular Neuroscience, HKUST, Clear Water Bay, Hong Kong, China
| | - Jianbo Yue
- Department of Biomedical Sciences, City University of Hong Kong, Kowloon Tong, Hong Kong, China.
| |
Collapse
|
31
|
Suzuki IK, Vanderhaeghen P. Is this a brain which I see before me? Modeling human neural development with pluripotent stem cells. Development 2016; 142:3138-50. [PMID: 26395142 DOI: 10.1242/dev.120568] [Citation(s) in RCA: 85] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
The human brain is arguably the most complex structure among living organisms. However, the specific mechanisms leading to this complexity remain incompletely understood, primarily because of the poor experimental accessibility of the human embryonic brain. Over recent years, technologies based on pluripotent stem cells (PSCs) have been developed to generate neural cells of various types. While the translational potential of PSC technologies for disease modeling and/or cell replacement therapies is usually put forward as a rationale for their utility, they are also opening novel windows for direct observation and experimentation of the basic mechanisms of human brain development. PSC-based studies have revealed that a number of cardinal features of neural ontogenesis are remarkably conserved in human models, which can be studied in a reductionist fashion. They have also revealed species-specific features, which constitute attractive lines of investigation to elucidate the mechanisms underlying the development of the human brain, and its link with evolution.
Collapse
Affiliation(s)
- Ikuo K Suzuki
- Université Libre de Bruxelles (ULB), Institute for Interdisciplinary Research (IRIBHM), and ULB Institute of Neuroscience (UNI), 808 Route de Lennik, Brussels B-1070, Belgium
| | - Pierre Vanderhaeghen
- Université Libre de Bruxelles (ULB), Institute for Interdisciplinary Research (IRIBHM), and ULB Institute of Neuroscience (UNI), 808 Route de Lennik, Brussels B-1070, Belgium WELBIO, Université Libre de Bruxelles, 808 Route de Lennik, Brussels B-1070, Belgium
| |
Collapse
|
32
|
Moreau M, Néant I, Webb SE, Miller AL, Riou JF, Leclerc C. Ca(2+) coding and decoding strategies for the specification of neural and renal precursor cells during development. Cell Calcium 2015; 59:75-83. [PMID: 26744233 DOI: 10.1016/j.ceca.2015.12.003] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2015] [Revised: 12/07/2015] [Accepted: 12/11/2015] [Indexed: 01/03/2023]
Abstract
During embryogenesis, a rise in intracellular Ca(2+) is known to be a widespread trigger for directing stem cells towards a specific tissue fate, but the precise Ca(2+) signalling mechanisms involved in achieving these pleiotropic effects are still poorly understood. In this review, we compare the Ca(2+) signalling events that appear to be one of the first steps in initiating and regulating both neural determination (neural induction) and kidney development (nephrogenesis). We have highlighted the necessary and sufficient role played by Ca(2+) influx and by Ca(2+) transients in the determination and differentiation of pools of neural or renal precursors. We have identified new Ca(2+) target genes involved in neural induction and we showed that the same Ca(2+) early target genes studied are not restricted to neural tissue but are also present in other tissues, principally in the pronephros. In this review, we also described a mechanism whereby the transcriptional control of gene expression during neurogenesis and nephrogenesis might be directly controlled by Ca(2+) signalling. This mechanism involves members of the Kcnip family such that a change in their binding properties to specific DNA sites is a result of Ca(2+) binding to EF-hand motifs. The different functions of Ca(2+) signalling during these two events illustrate the versatility of Ca(2+) as a second messenger.
Collapse
Affiliation(s)
- Marc Moreau
- Université Toulouse 3, Centre de Biologie du Développement, 118 route de Narbonne, F31062 Toulouse Cedex 04, France; CNRS UMR5547, Toulouse F31062, France
| | - Isabelle Néant
- Université Toulouse 3, Centre de Biologie du Développement, 118 route de Narbonne, F31062 Toulouse Cedex 04, France; CNRS UMR5547, Toulouse F31062, France
| | - Sarah E Webb
- Division of Life Science & State Key Laboratory of Molecular Neuroscience, HKUST, Clear Water Bay, Hong Kong, People's Republic of China
| | - Andrew L Miller
- Division of Life Science & State Key Laboratory of Molecular Neuroscience, HKUST, Clear Water Bay, Hong Kong, People's Republic of China; MBL, Woods Hole, MA, USA
| | - Jean-François Riou
- Université Pierre et Marie Curie-Paris VI, Equipe "Signalisation et Morphogenèse", UMR7622-Biologie du Développement, 9, quai Saint-Bernard, 75005 Paris, France; CNRS, Equipe "Signalisation et Morphogenèse", UMR7622-Biologie du Développement, 9, quai Saint-Bernard, 75005 Paris, France
| | - Catherine Leclerc
- Université Toulouse 3, Centre de Biologie du Développement, 118 route de Narbonne, F31062 Toulouse Cedex 04, France; CNRS UMR5547, Toulouse F31062, France.
| |
Collapse
|
33
|
Lim CS, Yang JE, Lee YK, Lee K, Lee JA, Kaang BK. Understanding the molecular basis of autism in a dish using hiPSCs-derived neurons from ASD patients. Mol Brain 2015; 8:57. [PMID: 26419846 PMCID: PMC4589208 DOI: 10.1186/s13041-015-0146-6] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2015] [Accepted: 09/11/2015] [Indexed: 12/20/2022] Open
Abstract
Autism spectrum disorder (ASD) is a complex neurodevelopmental disorder characterized by deficits in social cognition, language development, and repetitive/restricted behaviors. Due to the complexity and heterogeneity of ASD and lack of a proper human cellular model system, the pathophysiological mechanism of ASD during the developmental process is largely unknown. However, recent progress in induced pluripotent stem cell (iPSC) technology as well as in vitro neural differentiation techniques have allowed us to functionally characterize neurons and analyze cortical development during neural differentiation. These technical advances will increase our understanding of the pathogenic mechanisms of heterogeneous ASD and help identify molecular biomarkers for patient stratification as well as personalized medicine. In this review, we summarize our current knowledge of iPSC generation, differentiation of specific neuronal subtypes from iPSCs, and phenotypic characterizations of human ASD patient-derived iPSC models. Finally, we discuss the current limitations of iPSC technology and future directions of ASD pathophysiology studies using iPSCs.
Collapse
Affiliation(s)
- Chae-Seok Lim
- Department of Biological Sciences, College of Natural Sciences, Seoul National University, Gwanangno 599, Seoul, Gwanak-gu, 151-747, Korea
| | - Jung-Eun Yang
- Department of Biological Sciences, College of Natural Sciences, Seoul National University, Gwanangno 599, Seoul, Gwanak-gu, 151-747, Korea
| | - You-Kyung Lee
- Department of Biological Sciences and Biotechnology, College of Life Science and NanoTechnology, Hannam University, Jeonmin-dong 461-6, Daejeon, Yuseong-gu, 305-811, Korea
| | - Kyungmin Lee
- Department of Anatomy, Kyungpook National University Graduate School of Medicine, Dongin-dong 2-101, Daegu, Jung-gu, 700-422, Korea
| | - Jin-A Lee
- Department of Biological Sciences and Biotechnology, College of Life Science and NanoTechnology, Hannam University, Jeonmin-dong 461-6, Daejeon, Yuseong-gu, 305-811, Korea.
| | - Bong-Kiun Kaang
- Department of Biological Sciences, College of Natural Sciences, Seoul National University, Gwanangno 599, Seoul, Gwanak-gu, 151-747, Korea.
| |
Collapse
|
34
|
Pellett S, Schwartz MP, Tepp WH, Josephson R, Scherf JM, Pier CL, Thomson JA, Murphy WL, Johnson EA. Human Induced Pluripotent Stem Cell Derived Neuronal Cells Cultured on Chemically-Defined Hydrogels for Sensitive In Vitro Detection of Botulinum Neurotoxin. Sci Rep 2015; 5:14566. [PMID: 26411797 PMCID: PMC4585966 DOI: 10.1038/srep14566] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2015] [Accepted: 09/01/2015] [Indexed: 12/19/2022] Open
Abstract
Botulinum neurotoxin (BoNT) detection provides a useful model for validating cell-based neurotoxicity screening approaches, as sensitivity is dependent on functionally competent neurons and clear quantitative endpoints are available for correlating results to approved animal testing protocols. Here, human induced pluripotent stem cell (iPSC)-derived neuronal cells were cultured on chemically-defined poly(ethylene glycol) (PEG) hydrogels formed by "thiol-ene" photopolymerization and tested as a cell-based neurotoxicity assay by determining sensitivity to active BoNT/A1. BoNT/A1 sensitivity was comparable to the approved in vivo mouse bioassay for human iPSC-derived neurons and neural stem cells (iPSC-NSCs) cultured on PEG hydrogels or treated tissue culture polystyrene (TCP) surfaces. However, maximum sensitivity for BoNT detection was achieved two weeks earlier for iPSC-NSCs that were differentiated and matured on PEG hydrogels compared to TCP. Therefore, chemically-defined synthetic hydrogels offer benefits over standard platforms when optimizing culture conditions for cell-based screening and achieve sensitivities comparable to an approved animal testing protocol.
Collapse
Affiliation(s)
- Sabine Pellett
- Department of Bacteriology, University of Wisconsin at Madison, Madison, Wisconsin, United States of America
| | - Michael P. Schwartz
- Department of Biomedical Engineering, University of Wisconsin at Madison, Madison, Wisconsin, United States of America
| | - William H. Tepp
- Department of Bacteriology, University of Wisconsin at Madison, Madison, Wisconsin, United States of America
| | - Richard Josephson
- MTI-GlobalStem, Inc. Gaithersburg, Maryland, United States of America
| | - Jacob M. Scherf
- Department of Bacteriology, University of Wisconsin at Madison, Madison, Wisconsin, United States of America
| | - Christina L. Pier
- Department of Bacteriology, University of Wisconsin at Madison, Madison, Wisconsin, United States of America
| | - James A. Thomson
- Department of Cell and Regenerative Biology, University of Wisconsin at Madison, Madison, Wisconsin, United States of America
- Morgridge Institute for Research, Madison, Wisconsin, United States of America
- Department of Molecular, Cellular, and Developmental Biology, University of California at Santa Barbara, Santa Barbara, California, United States of America
| | - William L. Murphy
- Department of Biomedical Engineering, University of Wisconsin at Madison, Madison, Wisconsin, United States of America
- Department of Orthopedics and Rehabilitation, University of Wisconsin at Madison, Madison, Wisconsin, United States of America
| | - Eric A. Johnson
- Department of Bacteriology, University of Wisconsin at Madison, Madison, Wisconsin, United States of America
| |
Collapse
|
35
|
Human pluripotent stem cell-derived neural constructs for predicting neural toxicity. Proc Natl Acad Sci U S A 2015; 112:12516-21. [PMID: 26392547 DOI: 10.1073/pnas.1516645112] [Citation(s) in RCA: 245] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
Human pluripotent stem cell-based in vitro models that reflect human physiology have the potential to reduce the number of drug failures in clinical trials and offer a cost-effective approach for assessing chemical safety. Here, human embryonic stem (ES) cell-derived neural progenitor cells, endothelial cells, mesenchymal stem cells, and microglia/macrophage precursors were combined on chemically defined polyethylene glycol hydrogels and cultured in serum-free medium to model cellular interactions within the developing brain. The precursors self-assembled into 3D neural constructs with diverse neuronal and glial populations, interconnected vascular networks, and ramified microglia. Replicate constructs were reproducible by RNA sequencing (RNA-Seq) and expressed neurogenesis, vasculature development, and microglia genes. Linear support vector machines were used to construct a predictive model from RNA-Seq data for 240 neural constructs treated with 34 toxic and 26 nontoxic chemicals. The predictive model was evaluated using two standard hold-out testing methods: a nearly unbiased leave-one-out cross-validation for the 60 training compounds and an unbiased blinded trial using a single hold-out set of 10 additional chemicals. The linear support vector produced an estimate for future data of 0.91 in the cross-validation experiment and correctly classified 9 of 10 chemicals in the blinded trial.
Collapse
|
36
|
A natural diarylheptanoid promotes neuronal differentiation via activating ERK and PI3K-Akt dependent pathways. Neuroscience 2015; 303:389-401. [DOI: 10.1016/j.neuroscience.2015.07.019] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2015] [Revised: 06/02/2015] [Accepted: 07/06/2015] [Indexed: 11/22/2022]
|
37
|
Mimura S, Suga M, Liu Y, Kinehara M, Yanagihara K, Ohnuma K, Nikawa H, Furue MK. Synergistic effects of FGF-2 and Activin A on early neural differentiation of human pluripotent stem cells. In Vitro Cell Dev Biol Anim 2015; 51:769-75. [PMID: 25898826 DOI: 10.1007/s11626-015-9909-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2015] [Accepted: 04/01/2015] [Indexed: 10/23/2022]
Abstract
Neural differentiation is an important target of human embryonic stem cells, which provide a source for cell-based therapy, developmental biology, and pharmaceutical research. Previous studies revealed that inhibition of the bone morphogenetic protein is required for neural induction from human embryonic stem cells. On the contrary, the functions of fibroblast growth factors and Activin/Nodal signaling are controversial. Fibroblast growth factor-2 and Activin/Nodal pathways exert divergent influences on human embryonic stem cell concerning the maintenance of both pluripotency and cellular differentiation. We hypothesized that the combination of fibroblast growth factor-2 and Activin A at various concentrations synergistically exerts diverse effects on cell differentiation. To determine the effects of fibroblast growth factor-2 and Activin A on cellular differentiation into neural lineages, we examined the expression of neural differentiation markers in human embryonic stem cells treated with fibroblast growth factor-2 and/or Activin A at various concentrations in a growth factor-defined serum-free medium in short-term culture. In this study, we provide evidence that fibroblast growth factor-2 and Activin A synergistically regulated the initiation of human embryonic stem cell differentiation into neural cell lineages even though human embryonic stem cells autonomously differentiate into neural cell lineages.
Collapse
Affiliation(s)
- Sumiyo Mimura
- Laboratory of Stem Cell Cultures, National Institute of Biomedical Innovation, Health and Nutrition, 7-6-8 Saito Asagi, Ibaraki, Osaka, 567-0085, Japan.,Department of Oral Biology & Engineering, Integrated Health Sciences, Institute of Biomedical & Health Sciences, Hiroshima University, Hiroshima, 734-8553, Japan
| | - Mika Suga
- Laboratory of Stem Cell Cultures, National Institute of Biomedical Innovation, Health and Nutrition, 7-6-8 Saito Asagi, Ibaraki, Osaka, 567-0085, Japan
| | - Yujung Liu
- Laboratory of Stem Cell Cultures, National Institute of Biomedical Innovation, Health and Nutrition, 7-6-8 Saito Asagi, Ibaraki, Osaka, 567-0085, Japan
| | - Masaki Kinehara
- Laboratory of Stem Cell Cultures, National Institute of Biomedical Innovation, Health and Nutrition, 7-6-8 Saito Asagi, Ibaraki, Osaka, 567-0085, Japan.,Department of Cellular and Molecular Biology, Basic Life Sciences, Institute of Biomedical & Health Sciences, Hiroshima University, Hiroshima, 734-8553, Japan
| | - Kana Yanagihara
- Laboratory of Stem Cell Cultures, National Institute of Biomedical Innovation, Health and Nutrition, 7-6-8 Saito Asagi, Ibaraki, Osaka, 567-0085, Japan
| | - Kiyoshi Ohnuma
- Department of Bioengineering, Nagaoka University of Technology, Niigata, 940-2188, Japan
| | - Hiroki Nikawa
- Department of Oral Biology & Engineering, Integrated Health Sciences, Institute of Biomedical & Health Sciences, Hiroshima University, Hiroshima, 734-8553, Japan
| | - Miho K Furue
- Laboratory of Stem Cell Cultures, National Institute of Biomedical Innovation, Health and Nutrition, 7-6-8 Saito Asagi, Ibaraki, Osaka, 567-0085, Japan.
| |
Collapse
|
38
|
Thompson LH, Björklund A. Reconstruction of brain circuitry by neural transplants generated from pluripotent stem cells. Neurobiol Dis 2015; 79:28-40. [PMID: 25913029 DOI: 10.1016/j.nbd.2015.04.003] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2015] [Revised: 04/09/2015] [Accepted: 04/15/2015] [Indexed: 12/15/2022] Open
Abstract
Pluripotent stem cells (embryonic stem cells, ESCs, and induced pluripotent stem cells, iPSCs) have the capacity to generate neural progenitors that are intrinsically patterned to undergo differentiation into specific neuronal subtypes and express in vivo properties that match the ones formed during normal embryonic development. Remarkable progress has been made in this field during recent years thanks to the development of more refined protocols for the generation of transplantable neuronal progenitors from pluripotent stem cells, and the access to new tools for tracing of neuronal connectivity and assessment of integration and function of grafted neurons. Recent studies in brains of neonatal mice or rats, as well as in rodent models of brain or spinal cord damage, have shown that ESC- or iPSC-derived neural progenitors can be made to survive and differentiate after transplantation, and that they possess a remarkable capacity to extend axons over long distances and become functionally integrated into host neural circuitry. Here, we summarize these recent developments in the perspective of earlier studies using intracerebral and intraspinal transplants of primary neurons derived from fetal brain, with special focus on the ability of human ESC- and iPSC-derived progenitors to reconstruct damaged neural circuitry in cortex, hippocampus, the nigrostriatal system and the spinal cord, and we discuss the intrinsic and extrinsic factors that determine the growth properties of the grafted neurons and their capacity to establish target-specific long-distance axonal connections in the damaged host brain.
Collapse
Affiliation(s)
- Lachlan H Thompson
- Florey Institute for Neuroscience and Mental Health, University of Melbourne, Royal Parade, Parkville, Victoria 3010, Australia
| | - Anders Björklund
- Wallenberg Neuroscience Center, Department of Experimental Medical Science, Lund University, S-22184 Lund, Sweden.
| |
Collapse
|
39
|
Corti S, Faravelli I, Cardano M, Conti L. Human pluripotent stem cells as tools for neurodegenerative and neurodevelopmental disease modeling and drug discovery. Expert Opin Drug Discov 2015; 10:615-29. [PMID: 25891144 DOI: 10.1517/17460441.2015.1037737] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
INTRODUCTION Although intensive efforts have been made, effective treatments for neurodegenerative and neurodevelopmental diseases have not been yet discovered. Possible reasons for this include the lack of appropriate disease models of human neurons and a limited understanding of the etiological and neurobiological mechanisms. Recent advances in pluripotent stem cell (PSC) research have now opened the path to the generation of induced pluripotent stem cells (iPSCs) starting from somatic cells, thus offering an unlimited source of patient-specific disease-relevant neuronal cells. AREAS COVERED In this review, the authors focus on the use of human PSC-derived cells in modeling neurological disorders and discovering of new drugs and provide their expert perspectives on the field. EXPERT OPINION The advent of human iPSC-based disease models has fuelled renewed enthusiasm and enormous expectations for insights of disease mechanisms and identification of more disease-relevant and novel molecular targets. Human PSCs offer a unique tool that is being profitably exploited for high-throughput screening (HTS) platforms. This process can lead to the identification and optimization of molecules/drugs and thus move forward new pharmacological therapies for a wide range of neurodegenerative and neurodevelopmental conditions. It is predicted that improvements in the production of mature neuronal subtypes, from patient-specific human-induced pluripotent stem cells and their adaptation to culture, to HTS platforms will allow the increased exploitation of human pluripotent stem cells in drug discovery programs.
Collapse
Affiliation(s)
- Stefania Corti
- University of Milan, Dino Ferrari Centre, Neuroscience Section, Department of Pathophysiology and Transplantation, Neurology Unit, IRCCS Foundation Ca'Granda Ospedale Maggiore Policlinico , via Francesco Sforza 35, Milan 20122 , Italy +39 02 55033817 ;
| | | | | | | |
Collapse
|
40
|
Gaspar D, Spanoudes K, Holladay C, Pandit A, Zeugolis D. Progress in cell-based therapies for tendon repair. Adv Drug Deliv Rev 2015; 84:240-56. [PMID: 25543005 DOI: 10.1016/j.addr.2014.11.023] [Citation(s) in RCA: 127] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2014] [Revised: 11/08/2014] [Accepted: 11/12/2014] [Indexed: 02/07/2023]
Abstract
The last decade has seen significant developments in cell therapies, based on permanently differentiated, reprogrammed or engineered stem cells, for tendon injuries and degenerative conditions. In vitro studies assess the influence of biophysical, biochemical and biological signals on tenogenic phenotype maintenance and/or differentiation towards tenogenic lineage. However, the ideal culture environment has yet to be identified due to the lack of standardised experimental setup and readout system. Bone marrow mesenchymal stem cells and tenocytes/dermal fibroblasts appear to be the cell populations of choice for clinical translation in equine and human patients respectively based on circumstantial, rather than on hard evidence. Collaborative, inter- and multi-disciplinary efforts are expected to provide clinically relevant and commercially viable cell-based therapies for tendon repair and regeneration in the years to come.
Collapse
Affiliation(s)
- Diana Gaspar
- Network of Excellence for Functional Biomaterials (NFB), National University of Ireland, Galway (NUI Galway), Galway, Ireland
| | - Kyriakos Spanoudes
- Network of Excellence for Functional Biomaterials (NFB), National University of Ireland, Galway (NUI Galway), Galway, Ireland
| | - Carolyn Holladay
- Network of Excellence for Functional Biomaterials (NFB), National University of Ireland, Galway (NUI Galway), Galway, Ireland
| | - Abhay Pandit
- Network of Excellence for Functional Biomaterials (NFB), National University of Ireland, Galway (NUI Galway), Galway, Ireland
| | - Dimitrios Zeugolis
- Network of Excellence for Functional Biomaterials (NFB), National University of Ireland, Galway (NUI Galway), Galway, Ireland.
| |
Collapse
|
41
|
Intracellular Calcium Measurements for Functional Characterization of Neuronal Phenotypes. Methods Mol Biol 2015; 1341:245-55. [PMID: 26126448 DOI: 10.1007/7651_2015_271] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
The central and peripheral nervous system is built by a network of many different neuronal phenotypes together with glial and other supporting cells. The repertoire of expressed receptors and secreted neurotransmitters and neuromodulators are unique for each single neuron leading to intracellular signaling cascades, many of them involving intracellular calcium signaling. Here we suggest the use of calcium signaling analysis upon specific agonist application to reliably identify neuronal phenotypes, being important not only for basic science, but also providing a reliable tool for functional characterization of cells prior to transplantation. Calcium imaging provides various cellular information including signaling amplitudes, cell localization, duration, and frequency. Microfluorimetry reveals a signal summarizing the entire population, and its use is indicated for high-throughput screening purposes.
Collapse
|
42
|
Stappert L, Roese-Koerner B, Brüstle O. The role of microRNAs in human neural stem cells, neuronal differentiation and subtype specification. Cell Tissue Res 2015; 359:47-64. [PMID: 25172833 PMCID: PMC4284387 DOI: 10.1007/s00441-014-1981-y] [Citation(s) in RCA: 75] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2014] [Accepted: 07/28/2014] [Indexed: 12/20/2022]
Abstract
The impressive neuronal diversity found within the nervous system emerges from a limited pool of neural progenitor cells that proceed through different gene expression programs to acquire distinct cell fates. Here, we review recent evidence indicating that microRNAs (miRNAs) are critically involved in conferring neural cell identities during neural induction, neuronal differentiation and subtype specification. Several studies have shown that miRNAs act in concert with other gene regulatory factors and genetic switches to regulate the spatial and temporal expression profiles of important cell fate determinants. So far, most studies addressing the role of miRNAs during neurogenesis were conducted using animal models. With the advent of human pluripotent stem cells and the possibility to differentiate these into neural stem cells, we now have the opportunity to study miRNAs in a human context. More insight into the impact of miRNA-based regulation during neural fate choice could in the end be exploited to develop new strategies for the generation of distinct human neuronal cell types.
Collapse
Affiliation(s)
- Laura Stappert
- Institute of Reconstructive Neurobiology LIFE & BRAIN Center, University of Bonn and Hertie Foundation, Sigmund-Freud-Straße 25, Bonn, 53127 Germany
| | - Beate Roese-Koerner
- Institute of Reconstructive Neurobiology LIFE & BRAIN Center, University of Bonn and Hertie Foundation, Sigmund-Freud-Straße 25, Bonn, 53127 Germany
| | - Oliver Brüstle
- Institute of Reconstructive Neurobiology LIFE & BRAIN Center, University of Bonn and Hertie Foundation, Sigmund-Freud-Straße 25, Bonn, 53127 Germany
| |
Collapse
|
43
|
Moris-Sanz M, Estacio-Gómez A, Álvarez-Rivero J, Díaz-Benjumea FJ. Specification of neuronal subtypes by different levels of Hunchback. Development 2014; 141:4366-74. [DOI: 10.1242/dev.113381] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
During the development of the central nervous system, neural progenitors generate an enormous number of distinct types of neuron and glial cells by asymmetric division. Intrinsic genetic programs define the combinations of transcription factors that determine the fate of each cell, but the precise mechanisms by which all these factors are integrated at the level of individual cells are poorly understood. Here, we analyzed the specification of the neurons in the ventral nerve cord of Drosophila that express Crustacean cardioactive peptide (CCAP). There are two types of CCAP neurons: interneurons and efferent neurons. We found that both are specified during the Hunchback temporal window of neuroblast 3-5, but are not sibling cells. Further, this temporal window generates two ganglion mother cells that give rise to four neurons, which can be identified by the expression of empty spiracles. We show that the expression of Hunchback in the neuroblast increases over time and provide evidence that the absolute levels of Hunchback expression specify the two different CCAP neuronal fates.
Collapse
Affiliation(s)
- Marta Moris-Sanz
- Centro de Biología Molecular-Severo Ochoa (CSIC-UAM), c./Nicolás Cabrera 1, Universidad Autónoma, Madrid 28049, Spain
| | - Alicia Estacio-Gómez
- Centro de Biología Molecular-Severo Ochoa (CSIC-UAM), c./Nicolás Cabrera 1, Universidad Autónoma, Madrid 28049, Spain
| | - Javier Álvarez-Rivero
- Centro de Biología Molecular-Severo Ochoa (CSIC-UAM), c./Nicolás Cabrera 1, Universidad Autónoma, Madrid 28049, Spain
| | - Fernando J. Díaz-Benjumea
- Centro de Biología Molecular-Severo Ochoa (CSIC-UAM), c./Nicolás Cabrera 1, Universidad Autónoma, Madrid 28049, Spain
| |
Collapse
|
44
|
Moya N, Cutts J, Gaasterland T, Willert K, Brafman DA. Endogenous WNT signaling regulates hPSC-derived neural progenitor cell heterogeneity and specifies their regional identity. Stem Cell Reports 2014; 3:1015-28. [PMID: 25458891 PMCID: PMC4264562 DOI: 10.1016/j.stemcr.2014.10.004] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2014] [Revised: 10/13/2014] [Accepted: 10/14/2014] [Indexed: 11/20/2022] Open
Abstract
Neural progenitor cells (NPCs) derived from human pluripotent stem cells (hPSCs) are a multipotent cell population that is capable of nearly indefinite expansion and subsequent differentiation into the various neuronal and supporting cell types that comprise the CNS. However, current protocols for differentiating NPCs toward neuronal lineages result in a mixture of neurons from various regions of the CNS. In this study, we determined that endogenous WNT signaling is a primary contributor to the heterogeneity observed in NPC cultures and neuronal differentiation. Furthermore, exogenous manipulation of WNT signaling during neural differentiation, through either activation or inhibition, reduces this heterogeneity in NPC cultures, thereby promoting the formation of regionally homogeneous NPC and neuronal cultures. The ability to manipulate WNT signaling to generate regionally specific NPCs and neurons will be useful for studying human neural development and will greatly enhance the translational potential of hPSCs for neural-related therapies. Heterogeneous endogenous WNT signaling regulates hPSC-derived neuronal diversity Endogenous WNT signaling specifies the regional identity of hPSC-derived neurons Exogenous WNT signaling leads to uniform neuronal cultures from hPSCs Effects of WNT signaling on neurogenesis are recapitulated in an hPSC-based system
Collapse
Affiliation(s)
- Noel Moya
- Department of Cellular and Molecular Medicine, Stem Cell Program, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093-0695, USA
| | - Josh Cutts
- Department of Cellular and Molecular Medicine, Stem Cell Program, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093-0695, USA; School of Biological and Health Systems Engineering, Arizona State University, Tempe, AZ 85287-9709, USA
| | - Terry Gaasterland
- UCSD and Scripps Institution of Oceanography, Scripps Genome Center, 9500 Gilman Drive, La Jolla, CA 92093-0202, USA
| | - Karl Willert
- Department of Cellular and Molecular Medicine, Stem Cell Program, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093-0695, USA.
| | - David A Brafman
- Department of Cellular and Molecular Medicine, Stem Cell Program, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093-0695, USA; School of Biological and Health Systems Engineering, Arizona State University, Tempe, AZ 85287-9709, USA.
| |
Collapse
|
45
|
Sterthaus O, Feutz AC, Zhang H, Pletscher F, Bruder E, Miny P, Lezzi G, De Geyter M, De Geyter C. Gene expression profiles of similarly derived human embryonic stem cell lines correlate with their distinct propensity to exit stemness and their different differentiation behavior in culture. Cell Reprogram 2014; 16:185-95. [PMID: 24811852 DOI: 10.1089/cell.2013.0089] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Four normal-karyotype human embryonic stem cell (hESC) lines were generated using the same protocol and maintained under identical conditions. Despite these precautions, gene expression patterns were found to be dissimilar among the four lines. The observed differences were typical of each cell line, correlated with their distinct propensity to exit stemness, created heterogeneity among the cells during cell line maintenance, and correlated with their altered capacity as a source of differentiated cells. The capacity of some cell lines to give rise to more, and more mature, neurons within comparable time frames of directed differentiation reflected the distinct proportions of cells already predifferentiated at the onset. These findings demonstrate that the subsequent stages of neural differentiation were altered both in a quantitative and timely fashion. As a consequence, cell lines with apparent better and quicker ability to produce neurons were actually the less capable of reproducing proper differentiation. Previous data suggested that cell lines able to generate more neurons faster would be more suitable to clinical application. Our analysis of the differentiation process strongly suggests the opposite. The spontaneous tendency to predifferentiate of any particular hESC line should be known because it clearly impacts further experimental results.
Collapse
Affiliation(s)
- Oliver Sterthaus
- 1 Clinic of Gynecological Endocrinology and Reproductive Medicine, University of Basel , CH-4031, Basel, Switzerland
| | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Thinking out of the dish: what to learn about cortical development using pluripotent stem cells. Trends Neurosci 2014; 37:334-42. [PMID: 24745669 DOI: 10.1016/j.tins.2014.03.005] [Citation(s) in RCA: 74] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2013] [Revised: 03/16/2014] [Accepted: 03/18/2014] [Indexed: 01/07/2023]
Abstract
The development of the cerebral cortex requires the tightly coordinated generation of dozens of neuronal subtypes that will populate specific layers and areas. Recent studies have revealed how pluripotent stem cells (PSC), whether of mouse or human origin, can differentiate into a wide range of cortical neurons in vitro, which can integrate appropriately into the brain following in vivo transplantation. These models are largely artificial but recapitulate a substantial fraction of the complex temporal and regional patterning events that occur during in vivo corticogenesis. Here, we review these findings with emphasis on the new perspectives that they have brought for understanding of cortical development, evolution, and diseases.
Collapse
|
47
|
From pluripotency to forebrain patterning: an in vitro journey astride embryonic stem cells. Cell Mol Life Sci 2014; 71:2917-30. [PMID: 24643740 PMCID: PMC4098049 DOI: 10.1007/s00018-014-1596-1] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2013] [Revised: 02/17/2014] [Accepted: 02/26/2014] [Indexed: 02/07/2023]
Abstract
Embryonic stem cells (ESCs) have been used extensively as in vitro models of neural development and disease, with special efforts towards their conversion into forebrain progenitors and neurons. The forebrain is the most complex brain region, giving rise to several fundamental structures, such as the cerebral cortex, the hypothalamus, and the retina. Due to the multiplicity of signaling pathways playing different roles at distinct times of embryonic development, the specification and patterning of forebrain has been difficult to study in vivo. Research performed on ESCs in vitro has provided a large body of evidence to complement work in model organisms, but these studies have often been focused more on cell type production than on cell fate regulation. In this review, we systematically reassess the current literature in the field of forebrain development in mouse and human ESCs with a focus on the molecular mechanisms of early cell fate decisions, taking into consideration the specific culture conditions, exogenous and endogenous molecular cues as described in the original studies. The resulting model of early forebrain induction and patterning provides a useful framework for further studies aimed at reconstructing forebrain development in vitro for basic research or therapy.
Collapse
|
48
|
The ciliary proteins Meckelin and Jouberin are required for retinoic acid-dependent neural differentiation of mouse embryonic stem cells. Differentiation 2014; 87:134-146. [PMID: 24613594 DOI: 10.1016/j.diff.2014.02.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2013] [Revised: 01/23/2014] [Accepted: 02/17/2014] [Indexed: 12/29/2022]
Abstract
The dysfunction of the primary cilium, a complex, evolutionarily conserved, organelle playing an important role in sensing and transducing cell signals, is the unifying pathogenetic mechanism of a growing number of diseases collectively termed "ciliopathies", typically characterized by multiorgan involvement. Developmental defects of the central nervous system (CNS) characterize a subset of ciliopathies showing clinical and genetic overlap, such as Joubert syndrome (JS) and Meckel syndrome (MS). Although several knock-out mice lacking a variety of ciliary proteins have shown the importance of primary cilia in the development of the brain and CNS-derived structures, developmental in vitro studies, extremely useful to unravel the role of primary cilia along the course of neural differentiation, are still missing. Mouse embryonic stem cells (mESCs) have been recently proven to mimic brain development, giving the unique opportunity to dissect the CNS differentiation process along its sequential steps. In the present study we show that mESCs express the ciliary proteins Meckelin and Jouberin in a developmentally-regulated manner, and that these proteins co-localize with acetylated tubulin labeled cilia located at the outer embryonic layer. Further, mESCs differentiating along the neuronal lineage activate the cilia-dependent sonic hedgehog signaling machinery, which is impaired in Meckelin knock-out cells but results unaffected in Jouberin-deficient mESCs. However, both lose the ability to acquire a neuronal phenotype. Altogether, these results demonstrate a pivotal role of Meckelin and Jouberin during embryonic neural specification and indicate mESCs as a suitable tool to investigate the developmental impact of ciliary proteins dysfunction.
Collapse
|
49
|
Hao B, Lu Y, Wang Q, Guo W, Cheung KH, Yue J. Role of STIM1 in survival and neural differentiation of mouse embryonic stem cells independent of Orai1-mediated Ca2+ entry. Stem Cell Res 2014; 12:452-466. [PMID: 24424349 DOI: 10.1016/j.scr.2013.12.005] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2013] [Revised: 12/10/2013] [Accepted: 12/17/2013] [Indexed: 11/18/2022] Open
Abstract
Store-operated Ca(2+) entry (SOCE) is an important Ca(2+) influx pathway in non-excitable cells. STIM1, an ER Ca(2+) sensor, and Orai1, a plasma membrane Ca(2+) selective channel, are the two essential components of the Ca(2+) release activated channel (CRAC) responsible for SOCE activity. Here we explored the role of STIM1 and Orai1 in neural differentiation of mouse embryonic stem (ES) cells. We found that STIM1 and Orai1 were expressed and functionally active in ES cells, and expressions of STIM1 and Orai1 were dynamically regulated during neural differentiation of mouse ES cells. STIM1 knockdown inhibited the differentiation of mouse ES cells into neural progenitors, neurons, and astrocytes. In addition, STIM1 knockdown caused severe cell death and markedly suppressed the proliferation of neural progenitors. Surprisingly, Orai1 knockdown had little effect on neural differentiation of mouse ES cells, but the neurons derived from Orai1 knockdown ES cells, like those from STIM1 knockdown cells, had defective SOCE. Taken together, our data indicate that STIM1 is involved in both early neural differentiation of ES cells and survival of early differentiated ES cells independent of Orai1-mediated SOCE.
Collapse
Affiliation(s)
- Baixia Hao
- Department of Physiology, University of Hong Kong, Hong Kong, China
| | - Yingying Lu
- Department of Physiology, University of Hong Kong, Hong Kong, China
| | - Qian Wang
- Department of Physiology, University of Hong Kong, Hong Kong, China
| | - Wenjing Guo
- Department of Physiology, University of Hong Kong, Hong Kong, China
| | - King-Ho Cheung
- Department of Physiology, University of Hong Kong, Hong Kong, China
| | - Jianbo Yue
- Department of Physiology, University of Hong Kong, Hong Kong, China.
| |
Collapse
|
50
|
Nagashima F, Suzuki IK, Shitamukai A, Sakaguchi H, Iwashita M, Kobayashi T, Tone S, Toida K, Vanderhaeghen P, Kosodo Y. Novel and robust transplantation reveals the acquisition of polarized processes by cortical cells derived from mouse and human pluripotent stem cells. Stem Cells Dev 2014; 23:2129-42. [PMID: 24325299 DOI: 10.1089/scd.2013.0251] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Current stem cell technologies have enabled the induction of cortical progenitors and neurons from embryonic stem cells (ESCs) and induced pluripotent stem cells in vitro. To understand the mechanisms underlying the acquisition of apico-basal polarity and the formation of processes associated with the stemness of cortical cells generated in monolayer culture, here, we developed a novel in utero transplantation system based on the moderate dissociation of adherens junctions in neuroepithelial tissue. This method enables (1) the incorporation of remarkably higher numbers of grafted cells and (2) quantitative morphological analyses at single-cell resolution, including time-lapse recording analyses. We then grafted cortical progenitors induced from mouse ESCs into the developing brain. Importantly, we revealed that the mode of process extension depends on the extrinsic apico-basal polarity of the host epithelial tissue, as well as on the intrinsic differentiation state of the grafted cells. Further, we successfully transplanted cortical progenitors induced from human ESCs, showing that our strategy enables investigation of the neurogenesis of human neural progenitors within the developing mouse cortex. Specifically, human cortical cells exhibit multiple features of radial migration. The robust transplantation method established here could be utilized both to uncover the missing gap between neurogenesis from ESCs and the tissue environment and as an in vivo model of normal and pathological human corticogenesis.
Collapse
|