1
|
Como CN, O'Rourke R, Winkler C, Mitchell D, Tran L, Lorberbaum D, Sussel L, Franco S, Siegenthaler J. Meningeal-derived retinoic acid regulates neurogenesis via suppression of Notch and Sox2. Cell Rep 2025; 44:115637. [PMID: 40310723 DOI: 10.1016/j.celrep.2025.115637] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Revised: 02/03/2025] [Accepted: 04/10/2025] [Indexed: 05/03/2025] Open
Abstract
The meninges act as a regulator of brain development by secreting ligands that act on neural cells to regulate neurogenesis and neuronal migration. Meningeal-derived retinoic acid (RA) promotes neocortical neural progenitor cell cycle exit; however, the underlying molecular mechanism is unknown. Here, we used spatial transcriptomics and profiling of retinoic acid receptor α (RARα) DNA binding in Foxc1-mutant embryos that lack meninges-derived signals to identify potential neurogenic transcriptional mechanisms of RA signaling in telencephalic neural progenitors. This identified upregulation of Sox2 and Notch pathway genes, and RARα binds to the Sox2ot promoter, a long noncoding RNA that regulates Sox2 expression. Our experiments using maternal RA treatment and in utero electroporation in Foxc1 mutants support that meningeal-derived RA promotes neurogenesis by suppressing Notch signaling, a progenitor self-renewal pathway. Our findings elucidate a previously unknown mechanism of how meningeal RA coordinates neocortical development and provide insight into how defects in meningeal development may cause neurodevelopmental disorders.
Collapse
Affiliation(s)
- Christina N Como
- University of Colorado Anschutz Medical Campus, Department of Pediatrics, Section of Developmental Biology, Aurora, CO 80045, USA; University of Colorado Anschutz Medical Campus, Neuroscience Graduate Program, Aurora, CO 80045, USA
| | - Rebecca O'Rourke
- University of Colorado Anschutz Medical Campus, Department of Pediatrics, Section of Developmental Biology, Aurora, CO 80045, USA
| | - Caitlin Winkler
- University of Colorado Anschutz Medical Campus, Department of Pediatrics, Section of Developmental Biology, Aurora, CO 80045, USA
| | - Danae Mitchell
- University of Colorado Anschutz Medical Campus, Department of Pediatrics, Section of Developmental Biology, Aurora, CO 80045, USA; University of Colorado Anschutz Medical Campus, Neuroscience Graduate Program, Aurora, CO 80045, USA
| | - Luuli Tran
- University of Colorado Anschutz Medical Campus, Department of Pediatrics, Section of Developmental Biology, Aurora, CO 80045, USA; University of Colorado Anschutz Medical Campus, Molecular Biology Graduate Program, Aurora, CO 80045, USA
| | - David Lorberbaum
- University of Colorado Anschutz Medical Campus, Department of Pediatrics, Barbara Davis Center for Childhood Diabetes, Aurora, CO 80045, USA; University of Michigan Medical School, Department of Pharmacology and Caswell Diabetes Institute, Ann Arbor, MI 48105, USA
| | - Lori Sussel
- University of Colorado Anschutz Medical Campus, Department of Pediatrics, Barbara Davis Center for Childhood Diabetes, Aurora, CO 80045, USA
| | - Santos Franco
- University of Colorado Anschutz Medical Campus, Department of Pediatrics, Section of Developmental Biology, Aurora, CO 80045, USA.
| | - Julie Siegenthaler
- University of Colorado Anschutz Medical Campus, Department of Pediatrics, Section of Developmental Biology, Aurora, CO 80045, USA; University of Colorado Anschutz Medical Campus, Neuroscience Graduate Program, Aurora, CO 80045, USA.
| |
Collapse
|
2
|
Barkan E, Duran M, Lammers N, Tresenrider A, Jackson D, Lee H, Haagen B, Saunders L, Abitua P, Kimelman D, Trapnell C. Embryo-scale single-cell chemical transcriptomics reveals dependencies between cell types and signaling pathways. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.04.03.646423. [PMID: 40235986 PMCID: PMC11996465 DOI: 10.1101/2025.04.03.646423] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 04/17/2025]
Abstract
Organogenesis is a highly organized process that is conserved across vertebrates and is heavily dependent on intercellular signaling to achieve cell type identity. We lack a comprehensive understanding of how developing cell types in each organ and tissue depend on developmental signaling pathways. To address this gap in knowledge, we captured the molecular consequences of inhibiting each of the seven major developmental signaling pathways in zebrafish, using large-scale whole embryo single cell RNA-seq from over two million cells. This approach allowed us to detect signaling pathway regulation even in very rare cell types. By focusing on the development of the pectoral fin, we uncovered two new cell types (distal mesenchyme and tenocytes) and multiple novel signaling dependencies during pectoral fin development. This resource serves as a valuable tool for investigators seeking to rapidly assess the role of the major signaling pathways during the formation of their tissue of interest.
Collapse
|
3
|
Vivi E, Di Benedetto B. Brain stars take the lead during critical periods of early postnatal brain development: relevance of astrocytes in health and mental disorders. Mol Psychiatry 2024; 29:2821-2833. [PMID: 38553540 PMCID: PMC11420093 DOI: 10.1038/s41380-024-02534-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Revised: 03/12/2024] [Accepted: 03/14/2024] [Indexed: 09/25/2024]
Abstract
In the brain, astrocytes regulate shape and functions of the synaptic and vascular compartments through a variety of released factors and membrane-bound proteins. An imbalanced astrocyte activity can therefore have drastic negative impacts on brain development, leading to the onset of severe pathologies. Clinical and pre-clinical studies show alterations in astrocyte cell number, morphology, molecular makeup and astrocyte-dependent processes in different affected brain regions in neurodevelopmental (ND) and neuropsychiatric (NP) disorders. Astrocytes proliferate, differentiate and mature during the critical period of early postnatal brain development, a time window of elevated glia-dependent regulation of a proper balance between synapse formation/elimination, which is pivotal in refining synaptic connectivity. Therefore, any intrinsic and/or extrinsic factors altering these processes during the critical period may result in an aberrant synaptic remodeling and onset of mental disorders. The peculiar bridging position of astrocytes between synaptic and vascular compartments further allows them to "compute" the brain state and consequently secrete factors in the bloodstream, which may serve as diagnostic biomarkers of distinct healthy or disease conditions. Here, we collect recent advancements regarding astrogenesis and astrocyte-mediated regulation of neuronal network remodeling during early postnatal critical periods of brain development, focusing on synapse elimination. We then propose alternative hypotheses for an involvement of aberrancies in these processes in the onset of ND and NP disorders. In light of the well-known differential prevalence of certain brain disorders between males and females, we also discuss putative sex-dependent influences on these neurodevelopmental events. From a translational perspective, understanding age- and sex-dependent astrocyte-specific molecular and functional changes may help to identify biomarkers of distinct cellular (dys)functions in health and disease, favouring the development of diagnostic tools or the selection of tailored treatment options for male/female patients.
Collapse
Affiliation(s)
- Eugenia Vivi
- Laboratory of Neuro-Glia Pharmacology, Department of Psychiatry and Psychotherapy, University of Regensburg, 93053, Regensburg, Germany
| | - Barbara Di Benedetto
- Laboratory of Neuro-Glia Pharmacology, Department of Psychiatry and Psychotherapy, University of Regensburg, 93053, Regensburg, Germany.
- Regensburg Center of Neuroscience, University of Regensburg, Regensburg, Germany.
| |
Collapse
|
4
|
Ma Z, Zeng Y, Wang M, Liu W, Zhou J, Wu C, Hou L, Yin B, Qiang B, Shu P, Peng X. N4BP1 mediates RAM domain-dependent notch signaling turnover during neocortical development. EMBO J 2023; 42:e113383. [PMID: 37807845 PMCID: PMC10646556 DOI: 10.15252/embj.2022113383] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2022] [Revised: 09/06/2023] [Accepted: 09/07/2023] [Indexed: 10/10/2023] Open
Abstract
Notch signaling pathway activity, particularly fluctuations in the biologically active effector fragment NICD, is required for rapid and efficient dynamic regulation of proper fate decisions in stem cells. In this study, we identified NEDD4-binding protein 1 (N4BP1), which is highly expressed in the developing mouse cerebral cortex, as a negative modulator of Notch signaling dynamics in neural progenitor cells. Intriguingly, N4BP1 regulated NICD stability specifically after Notch1 S3 cleavage through ubiquitin-mediated degradation that depended on its RAM domain, not its PEST domain, as had been extensively and previously described. The CoCUN domain in N4BP1, particularly the "Phe-Pro" motif (862/863 amino acid), was indispensable for mediating NICD degradation. The Ring family E3 ligase Trim21 was, in contrast to other NEDD4 family members, required for N4BP1-regulated NICD degradation. Overexpression of N4BP1 in cortical neural progenitors promoted neural stem cell differentiation, whereas neural progenitor cells lacking N4BP1 were sensitized to Notch signaling, resulting in the maintenance of stem-like properties in neural progenitor cells and lower production of cortical neurons.
Collapse
Affiliation(s)
- Zhihua Ma
- Department of Molecular Biology and Biochemistry, Medical Primate Research Center, Neuroscience CenterInstitute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical CollegeBeijingChina
| | - Yi Zeng
- Department of Molecular Biology and Biochemistry, Medical Primate Research Center, Neuroscience CenterInstitute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical CollegeBeijingChina
- Present address:
Department of Infectious Diseases, Institute for Viral Hepatitis, The Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education)The Second Affiliated Hospital of Chongqing Medical UniversityChongqingChina
| | - Ming Wang
- Department of Molecular Biology and Biochemistry, Medical Primate Research Center, Neuroscience CenterInstitute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical CollegeBeijingChina
- Present address:
Department of Otolaryngology, Head and Neck Surgery, Beijing Tongren HospitalCapital Medical University, Beijing Key Laboratory of Nasal Diseases, Beijing Institute of OtolaryngologyBeijingChina
| | - Wei Liu
- Department of Molecular Biology and Biochemistry, Medical Primate Research Center, Neuroscience CenterInstitute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical CollegeBeijingChina
| | - Jiafeng Zhou
- Department of Molecular Biology and Biochemistry, Medical Primate Research Center, Neuroscience CenterInstitute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical CollegeBeijingChina
| | - Chao Wu
- Department of Molecular Biology and Biochemistry, Medical Primate Research Center, Neuroscience CenterInstitute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical CollegeBeijingChina
| | - Lin Hou
- Department of Molecular Biology and Biochemistry, Medical Primate Research Center, Neuroscience CenterInstitute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical CollegeBeijingChina
- State Key Laboratory of Common Mechanism Research for Major DiseasesBeijingChina
| | - Bin Yin
- Department of Molecular Biology and Biochemistry, Medical Primate Research Center, Neuroscience CenterInstitute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical CollegeBeijingChina
- State Key Laboratory of Common Mechanism Research for Major DiseasesBeijingChina
| | - Boqin Qiang
- Department of Molecular Biology and Biochemistry, Medical Primate Research Center, Neuroscience CenterInstitute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical CollegeBeijingChina
- State Key Laboratory of Common Mechanism Research for Major DiseasesBeijingChina
| | - Pengcheng Shu
- Department of Molecular Biology and Biochemistry, Medical Primate Research Center, Neuroscience CenterInstitute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical CollegeBeijingChina
- State Key Laboratory of Common Mechanism Research for Major DiseasesBeijingChina
- Chinese Institute for Brain ResearchBeijingChina
| | - Xiaozhong Peng
- Department of Molecular Biology and Biochemistry, Medical Primate Research Center, Neuroscience CenterInstitute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical CollegeBeijingChina
- State Key Laboratory of Respiratory Health and MultimorbidityBeijingChina
- Institute of Laboratory Animal ScienceChinese Academy of Medical Sciences, Peking Union Medical CollegeBeijingChina
| |
Collapse
|
5
|
Martin M, Gutierrez-Avino F, Shaikh MN, Tejedor FJ. A novel proneural function of Asense is integrated with the sequential actions of Delta-Notch, L'sc and Su(H) to promote the neuroepithelial to neuroblast transition. PLoS Genet 2023; 19:e1010991. [PMID: 37871020 PMCID: PMC10621995 DOI: 10.1371/journal.pgen.1010991] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2023] [Revised: 11/02/2023] [Accepted: 09/20/2023] [Indexed: 10/25/2023] Open
Abstract
In order for neural progenitors (NPs) to generate distinct populations of neurons at the right time and place during CNS development, they must switch from undergoing purely proliferative, self-renewing divisions to neurogenic, asymmetric divisions in a tightly regulated manner. In the developing Drosophila optic lobe, neuroepithelial (NE) cells of the outer proliferation center (OPC) are progressively transformed into neurogenic NPs called neuroblasts (NBs) in a medial to lateral proneural wave. The cells undergoing this transition express Lethal of Scute (L'sc), a proneural transcription factor (TF) of the Acheate Scute Complex (AS-C). Here we show that there is also a peak of expression of Asense (Ase), another AS-C TF, in the cells neighboring those with transient L'sc expression. These peak of Ase cells help to identify a new transitional stage as they have lost NE markers and L'sc, they receive a strong Notch signal and barely exhibit NB markers. This expression of Ase is necessary and sufficient to promote the NE to NB transition in a more robust and rapid manner than that of l'sc gain of function or Notch loss of function. Thus, to our knowledge, these data provide the first direct evidence of a proneural role for Ase in CNS neurogenesis. Strikingly, we found that strong Delta-Notch signaling at the lateral border of the NE triggers l'sc expression, which in turn induces ase expression in the adjacent cells through the activation of Delta-Notch signaling. These results reveal two novel non-conventional actions of Notch signaling in driving the expression of proneural factors, in contrast to the repression that Notch signaling exerts on them during classical lateral inhibition. Finally, Suppressor of Hairless (Su(H)), which seems to be upregulated late in the transitioning cells and in NBs, represses l'sc and ase, ensuring their expression is transient. Thus, our data identify a key proneural role of Ase that is integrated with the sequential activities of Delta-Notch signaling, L'sc, and Su(H), driving the progressive transformation of NE cells into NBs.
Collapse
Affiliation(s)
- Mercedes Martin
- Instituto de Neurociencias, Consejo Superior de Investigaciones Científicas and Universidad Miguel Hernandez, Sant Joan d’Alacant, Spain
| | - Francisco Gutierrez-Avino
- Instituto de Neurociencias, Consejo Superior de Investigaciones Científicas and Universidad Miguel Hernandez, Sant Joan d’Alacant, Spain
| | - Mirja N. Shaikh
- Instituto de Neurociencias, Consejo Superior de Investigaciones Científicas and Universidad Miguel Hernandez, Sant Joan d’Alacant, Spain
| | - Francisco J. Tejedor
- Instituto de Neurociencias, Consejo Superior de Investigaciones Científicas and Universidad Miguel Hernandez, Sant Joan d’Alacant, Spain
| |
Collapse
|
6
|
Yang C, Shitamukai A, Yang S, Kawaguchi A. Advanced Techniques Using In Vivo Electroporation to Study the Molecular Mechanisms of Cerebral Development Disorders. Int J Mol Sci 2023; 24:14128. [PMID: 37762431 PMCID: PMC10531473 DOI: 10.3390/ijms241814128] [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: 08/10/2023] [Revised: 09/12/2023] [Accepted: 09/13/2023] [Indexed: 09/29/2023] Open
Abstract
The mammalian cerebral cortex undergoes a strictly regulated developmental process. Detailed in situ visualizations, imaging of these dynamic processes, and in vivo functional gene studies significantly enhance our understanding of brain development and related disorders. This review introduces basic techniques and recent advancements in in vivo electroporation for investigating the molecular mechanisms underlying cerebral diseases. In utero electroporation (IUE) is extensively used to visualize and modify these processes, including the forced expression of pathological mutants in human diseases; thus, this method can be used to establish animal disease models. The advent of advanced techniques, such as genome editing, including de novo knockout, knock-in, epigenetic editing, and spatiotemporal gene regulation, has further expanded our list of investigative tools. These tools include the iON expression switch for the precise control of timing and copy numbers of exogenous genes and TEMPO for investigating the temporal effects of genes. We also introduce the iGONAD method, an improved genome editing via oviductal nucleic acid delivery approach, as a novel genome-editing technique that has accelerated brain development exploration. These advanced in vivo electroporation methods are expected to provide valuable insights into pathological conditions associated with human brain disorders.
Collapse
Affiliation(s)
- Chen Yang
- Human Anatomy and Histology and Embryology, School of Basic Medicine, Harbin Medical University, Harbin 150081, China
- Department of Human Morphology, Okayama University Graduate School of Medicine, Density and Pharmaceutical Sciences, Okayama 700-8558, Japan
| | - Atsunori Shitamukai
- Department of Human Morphology, Okayama University Graduate School of Medicine, Density and Pharmaceutical Sciences, Okayama 700-8558, Japan
| | - Shucai Yang
- Human Anatomy and Histology and Embryology, School of Basic Medicine, Harbin Medical University, Harbin 150081, China
| | - Ayano Kawaguchi
- Department of Human Morphology, Okayama University Graduate School of Medicine, Density and Pharmaceutical Sciences, Okayama 700-8558, Japan
| |
Collapse
|
7
|
Assessment of Dynein-Mediated Nuclear Migration in the Developing Cortex by Live-Tissue Microscopy. Methods Mol Biol 2023; 2623:61-71. [PMID: 36602679 DOI: 10.1007/978-1-0716-2958-1_4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
During development of the cerebral cortex, neuroepithelial and radial glial cells undergo an oscillatory nuclear movement throughout their cell cycle, termed interkinetic nuclear migration. The nucleus of postmitotic neurons derived from these neural stem cells also translocates in a saltatory manner to enable neuronal migration toward the cortical plate. In these processes, various molecular motors, including cytoplasmic dynein, myosin II, and kinesins, are the driving force for nuclear migration at different stages. Despite efforts made to understand the mechanism regulating cortical development over decades, novel gene mutations discovered in neurodevelopmental disorders indicate that missing pieces still remain. Gene manipulation by in utero electroporation combined with live microscopy of neural stem cells in brain slices provides a powerful method to capture their detailed behaviors during proliferation and migration. The procedures described in this chapter enable the monitoring of cell cycle progression, mitosis, morphological changes, and migratory patterns in situ. This approach facilitates the elucidation of gene functions in cortical development and neurodevelopmental disorders.
Collapse
|
8
|
Liu JW, Li H, Zhang Y. Npas3 regulates stemness maintenance of radial glial cells and neuronal migration in the developing mouse cerebral cortex. Front Cell Neurosci 2022; 16:865681. [PMID: 36313621 PMCID: PMC9608153 DOI: 10.3389/fncel.2022.865681] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Accepted: 09/20/2022] [Indexed: 11/13/2022] Open
Abstract
The neuronal PAS domain 3 (NPAS3) is a member of the basic helix-loop-helix (bHLH) PAS family of transcription factors and is implicated in psychiatric and neurodevelopmental disorders. NPAS3 is robustly expressed in the cortical ventricle zone (VZ), a transient proliferative zone containing progenitor cells, mainly radial glial cells, destined to give rise to cortical excitatory neurons. However, the role of NPAS3 in corticogenesis remains largely unknown. In this study, we knocked down Npas3 expression in the neural progenitor cells residing in the cortical VZ to investigate the role of Npas3 in cerebral cortical development in mice. We demonstrated that Npas3 knockdown profoundly impaired neuronal radial migration and changed the laminar cell fate of the cells detained in the deep cortical layers. Furthermore, the downregulation of Npas3 led to the stemness maintenance of radial glial cells and increased the proliferation rate of neural progenitor cells residing in the VZ/subventricular zone (SVZ). These findings underline the function of Npas3 in the development of the cerebral cortex and may shed light on the etiology of NPAS3-related disorders.
Collapse
|
9
|
Coelho P, Fão L, Mota S, Rego AC. Mitochondrial function and dynamics in neural stem cells and neurogenesis: Implications for neurodegenerative diseases. Ageing Res Rev 2022; 80:101667. [PMID: 35714855 DOI: 10.1016/j.arr.2022.101667] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Revised: 05/21/2022] [Accepted: 06/09/2022] [Indexed: 11/28/2022]
Abstract
Mitochondria have been largely described as the powerhouse of the cell and recent findings demonstrate that this organelle is fundamental for neurogenesis. The mechanisms underlying neural stem cells (NSCs) maintenance and differentiation are highly regulated by both intrinsic and extrinsic factors. Mitochondrial-mediated switch from glycolysis to oxidative phosphorylation, accompanied by mitochondrial remodeling and dynamics are vital to NSCs fate. Deregulation of mitochondrial proteins, mitochondrial DNA, function, fission/fusion and metabolism underly several neurodegenerative diseases; data show that these impairments are already present in early developmental stages and NSC fate decisions. However, little is known about mitochondrial role in neurogenesis. In this Review, we describe the recent evidence covering mitochondrial role in neurogenesis, its impact in selected neurodegenerative diseases, for which aging is the major risk factor, and the recent advances in stem cell-based therapies that may alleviate neurodegenerative disorders-related neuronal deregulation through improvement of mitochondrial function and dynamics.
Collapse
Affiliation(s)
- Patrícia Coelho
- CNC, Center for Neuroscience and Cell Biology, University of Coimbra Polo 1, Coimbra, Portugal.
| | - Lígia Fão
- CNC, Center for Neuroscience and Cell Biology, University of Coimbra Polo 1, Coimbra, Portugal; FMUC- Faculty of Medicine, University of Coimbra Polo 3, Coimbra, Portugal.
| | - Sandra Mota
- CNC, Center for Neuroscience and Cell Biology, University of Coimbra Polo 1, Coimbra, Portugal; III, Institute of Interdisciplinary Research, University of Coimbra, Coimbra, Portugal.
| | - A Cristina Rego
- CNC, Center for Neuroscience and Cell Biology, University of Coimbra Polo 1, Coimbra, Portugal; FMUC- Faculty of Medicine, University of Coimbra Polo 3, Coimbra, Portugal.
| |
Collapse
|
10
|
Reduced chromatin accessibility correlates with resistance to Notch activation. Nat Commun 2022; 13:2210. [PMID: 35468895 PMCID: PMC9039071 DOI: 10.1038/s41467-022-29834-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Accepted: 03/18/2022] [Indexed: 11/08/2022] Open
Abstract
The Notch signalling pathway is a master regulator of cell fate transitions in development and disease. In the brain, Notch promotes neural stem cell (NSC) proliferation, regulates neuronal migration and maturation and can act as an oncogene or tumour suppressor. How NOTCH and its transcription factor RBPJ activate distinct gene regulatory networks in closely related cell types in vivo remains to be determined. Here we use Targeted DamID (TaDa), requiring only thousands of cells, to identify NOTCH and RBPJ binding in NSCs and their progeny in the mouse embryonic cerebral cortex in vivo. We find that NOTCH and RBPJ associate with a broad network of NSC genes. Repression of NSC-specific Notch target genes in intermediate progenitors and neurons correlates with decreased chromatin accessibility, suggesting that chromatin compaction may contribute to restricting NOTCH-mediated transactivation.
Collapse
|
11
|
Wu Q, Shichino Y, Abe T, Suetsugu T, Omori A, Kiyonari H, Iwasaki S, Matsuzaki F. Selective translation of epigenetic modifiers affects the temporal pattern and differentiation of neural stem cells. Nat Commun 2022; 13:470. [PMID: 35078993 PMCID: PMC8789897 DOI: 10.1038/s41467-022-28097-y] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Accepted: 01/07/2022] [Indexed: 11/09/2022] Open
Abstract
The cerebral cortex is formed by diverse neurons generated sequentially from neural stem cells (NSCs). A clock mechanism has been suggested to underlie the temporal progression of NSCs, which is mainly defined by the transcriptome and the epigenetic state. However, what drives such a developmental clock remains elusive. We show that translational control of histone H3 trimethylation in Lys27 (H3K27me3) modifiers is part of this clock. We find that depletion of Fbl, an rRNA methyltransferase, reduces translation of both Ezh2 methyltransferase and Kdm6b demethylase of H3K27me3 and delays the progression of the NSC state. These defects are partially phenocopied by simultaneous inhibition of H3K27me3 methyltransferase and demethylase, indicating the role of Fbl in the genome-wide H3K27me3 pattern. Therefore, we propose that Fbl drives the intrinsic clock through the translational enhancement of the H3K27me3 modifiers that predominantly define the NSC state. The temporal development of tissues and organs may be defined by the genome-wide epigenetic and transcriptional state functioning as the clock. Here the authors found that Fbl, a ribosomal RNA methyltransferase, potentially behaves as a clock during neural stem cell (NSC) development by controlling translational efficiencies of epigenetic modifiers in the cerebral cortex primordium.
Collapse
|
12
|
Temperature sensitivity of Notch signaling underlies species-specific developmental plasticity and robustness in amniote brains. Nat Commun 2022; 13:96. [PMID: 35013223 PMCID: PMC8748702 DOI: 10.1038/s41467-021-27707-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2020] [Accepted: 12/06/2021] [Indexed: 11/08/2022] Open
Abstract
Ambient temperature significantly affects developmental timing in animals. The temperature sensitivity of embryogenesis is generally believed to be a consequence of the thermal dependency of cellular metabolism. However, the adaptive molecular mechanisms that respond to variations in temperature remain unclear. Here, we report species-specific thermal sensitivity of Notch signaling in the developing amniote brain. Transient hypothermic conditions increase canonical Notch activity and reduce neurogenesis in chick neural progenitors. Increased biosynthesis of phosphatidylethanolamine, a major glycerophospholipid components of the plasma membrane, mediates hypothermia-induced Notch activation. Furthermore, the species-specific thermal dependency of Notch signaling is associated with developmental robustness to altered Notch signaling. Our results reveal unique regulatory mechanisms for temperature-dependent neurogenic potentials that underlie developmental and evolutionary adaptations to a range of ambient temperatures in amniotes. Ambient temperature significantly affects embryogenesis, but adaptive molecular mechanisms that respond to temperature remain unclear. Here, the authors identified species-specific thermal sensitivity of Notch signaling in developing amniote brains.
Collapse
|
13
|
Mizutani KI. [Spatiotemporally Dependent Vascularization Regulates Neural Stem and Progenitor Cells]. YAKUGAKU ZASSHI 2021; 141:335-341. [PMID: 33642501 DOI: 10.1248/yakushi.20-00198-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Blood vessels including arteries, veins, and capillaries, are densely spread throughout the body. One round of systemic blood circulation through these blood vessels occurs approximately every minute, and blood sent by the heart transports oxygen, nutrients, and fluid to cells throughout the body. This nourishes cells, tissues, and organs and maintains homeostasis. The relatively simple structure of blood vessels consists of endothelial cells surrounded by a basal lamina and pericytes covering the outer layer. However, blood vessels patterning markedly varies among tissues. The diversity and plasticity of vascular networks are considered vital for this system to facilitate distinct functions for each tissue. Recent studies revealed that blood vessels create a tissue-specific niche, thus attracting attention as biologically active sites for tissue development. This vascular niche establishes specialized microenvironments through both direct physical contact and secreted-soluble factors. Here, we review advances in our understanding of how the vascular niche is utilized by neural stem and progenitor cells during neocortical development, and describe future perspectives regarding new treatment strategies for neural diseases utilizing this vascular niche.
Collapse
Affiliation(s)
- Ken-Ichi Mizutani
- Laboratory of Stem Cell Biology, Graduate School of Pharmaceutical Sciences, Kobe Gakuin University
| |
Collapse
|
14
|
Hatakeyama J, Shimamura K. The Pace of Neurogenesis Is Regulated by the Transient Retention of the Apical Endfeet of Differentiating Cells. Cereb Cortex 2020; 29:3725-3737. [PMID: 30307484 DOI: 10.1093/cercor/bhy252] [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: 12/09/2015] [Revised: 09/08/2018] [Accepted: 09/13/2018] [Indexed: 01/08/2023] Open
Abstract
The development of the mammalian cerebral cortex involves a variety of temporally organized events such as successive waves of neuronal production and the transition of progenitor competence for each neuronal subtype generated. The number of neurons generated in a certain time period, that is, the rate of neuron production, varies across the regions of the brain and the specific developmental stage; however, the underlying mechanism of this process is poorly understood. We have recently found that nascent neurons communicate with undifferentiated progenitors and thereby regulate neurogenesis, through a transiently retained apical endfoot that signals via the Notch pathway. Here, we report that the retention time length of the neuronal apical endfoot correlates with the rate of neuronal production in the developing mouse cerebral cortex. We further demonstrate that a forced reduction or extension of the retention period through the disruption or stabilization of adherens junction, respectively, resulted in the acceleration or deceleration of neurogenesis, respectively. Our results suggest that the apical endfeet of differentiating cells serve as a pace controller for neurogenesis, thereby assuring the well-proportioned laminar organization of the neocortex.
Collapse
Affiliation(s)
- Jun Hatakeyama
- Department of Brain Morphogenesis, Institute of Molecular Embryology and Genetics, Kumamoto University, 2-2-1 Honjo, Chuo-ku, Kumamoto, Japan
| | - Kenji Shimamura
- Department of Brain Morphogenesis, Institute of Molecular Embryology and Genetics, Kumamoto University, 2-2-1 Honjo, Chuo-ku, Kumamoto, Japan
| |
Collapse
|
15
|
Hagey DW, Topcic D, Kee N, Reynaud F, Bergsland M, Perlmann T, Muhr J. CYCLIN-B1/2 and -D1 act in opposition to coordinate cortical progenitor self-renewal and lineage commitment. Nat Commun 2020; 11:2898. [PMID: 32518258 PMCID: PMC7283355 DOI: 10.1038/s41467-020-16597-8] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Accepted: 05/13/2020] [Indexed: 11/09/2022] Open
Abstract
The sequential generation of layer-specific cortical neurons requires radial glia cells (RGCs) to precisely balance self-renewal and lineage commitment. While specific cell-cycle phases have been associated with these decisions, the mechanisms linking the cell-cycle machinery to cell-fate commitment remain obscure. Using single-cell RNA-sequencing, we find that the strongest transcriptional signature defining multipotent RGCs is that of G2/M-phase, and particularly CYCLIN-B1/2, while lineage-committed progenitors are enriched in G1/S-phase genes, including CYCLIN-D1. These data also reveal cell-surface markers that allow us to isolate RGCs and lineage-committed progenitors, and functionally confirm the relationship between cell-cycle phase enrichment and cell fate competence. Finally, we use cortical electroporation to demonstrate that CYCLIN-B1/2 cooperate with CDK1 to maintain uncommitted RGCs by activating the NOTCH pathway, and that CYCLIN-D1 promotes differentiation. Thus, this work establishes that cell-cycle phase-specific regulators act in opposition to coordinate the self-renewal and lineage commitment of RGCs via core stem cell regulatory pathways.
Collapse
Affiliation(s)
- Daniel W Hagey
- Department of Cell and Molecular Biology, Karolinska Institutet, Solnavägen 9, SE-171 65, Stockholm, Sweden.
| | - Danijal Topcic
- Department of Cell and Molecular Biology, Karolinska Institutet, Solnavägen 9, SE-171 65, Stockholm, Sweden
| | - Nigel Kee
- Department of Cell and Molecular Biology, Karolinska Institutet, Solnavägen 9, SE-171 65, Stockholm, Sweden
| | - Florie Reynaud
- Department of Cell and Molecular Biology, Karolinska Institutet, Solnavägen 9, SE-171 65, Stockholm, Sweden
| | - Maria Bergsland
- Department of Cell and Molecular Biology, Karolinska Institutet, Solnavägen 9, SE-171 65, Stockholm, Sweden
| | - Thomas Perlmann
- Department of Cell and Molecular Biology, Karolinska Institutet, Solnavägen 9, SE-171 65, Stockholm, Sweden
| | - Jonas Muhr
- Department of Cell and Molecular Biology, Karolinska Institutet, Solnavägen 9, SE-171 65, Stockholm, Sweden.
| |
Collapse
|
16
|
Esteve P, Crespo I, Kaimakis P, Sandonís A, Bovolenta P. Sfrp1 Modulates Cell-signaling Events Underlying Telencephalic Patterning, Growth and Differentiation. Cereb Cortex 2020; 29:1059-1074. [PMID: 30084950 DOI: 10.1093/cercor/bhy013] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2017] [Accepted: 01/09/2018] [Indexed: 12/19/2022] Open
Abstract
The mammalian dorsal telencephalic neuroepithelium develops-from medial to lateral-into the choroid plaque, cortical hem, hippocampal primordium and isocortex under the influence of Bmp, Wnt and Notch signaling. Correct telencephalic development requires a tight coordination of the extent/duration of these signals, but the identification of possible molecular coordinators is still limited. Here, we postulated that Secreted Frizzled Related Protein 1 (Sfrp1), a multifunctional regulator of Bmp, Wnt and Notch signaling strongly expressed during early telencephalic development, may represent 1 of such molecules. We report that in E10.5-E12.5 Sfrp1-/- embryos, the hem and hippocampal domains are reduced in size whereas the prospective neocortex is medially extended. These changes are associated with a significant reduction of the medio-lateral telencephalic expression of Axin2, a read-out of Wnt/βcatenin signaling activation. Furthermore, in the absence of Sfrp1, Notch signaling is increased, cortical progenitor cell cycle is shorter, with expanded progenitor pools and enhanced generation of early-born neurons. Hence, in postnatal Sfrp1-/- animals the anterior hippocampus is reduced and the neocortex is shorter in the antero-posterior and medio-lateral axis but is thicker. We propose that, by controlling Wnt and Notch signaling in opposite directions, Sfrp1 promotes hippocampal patterning and balances medio-lateral and antero-posterior cortex expansion.
Collapse
Affiliation(s)
- Pilar Esteve
- Centro de Biología Molecular "Severo Ochoa", CSIC-UAM and CIBER de Enfermedades Raras (CIBERER), c/Nicolás Cabrera, Madrid, Spain
| | - Inmaculada Crespo
- Centro de Biología Molecular "Severo Ochoa", CSIC-UAM and CIBER de Enfermedades Raras (CIBERER), c/Nicolás Cabrera, Madrid, Spain
| | - Polynikis Kaimakis
- Centro de Biología Molecular "Severo Ochoa", CSIC-UAM and CIBER de Enfermedades Raras (CIBERER), c/Nicolás Cabrera, Madrid, Spain
| | - Africa Sandonís
- Centro de Biología Molecular "Severo Ochoa", CSIC-UAM and CIBER de Enfermedades Raras (CIBERER), c/Nicolás Cabrera, Madrid, Spain
| | - Paola Bovolenta
- Centro de Biología Molecular "Severo Ochoa", CSIC-UAM and CIBER de Enfermedades Raras (CIBERER), c/Nicolás Cabrera, Madrid, Spain
| |
Collapse
|
17
|
Wang B, Ji L, Bishayee K, Li C, Huh SO. Identification and prevention of heterotopias in mouse neocortical neural cell migration incurred by surgical damages during utero electroporation procedures. Anim Cells Syst (Seoul) 2020; 24:114-123. [PMID: 32489691 PMCID: PMC7241496 DOI: 10.1080/19768354.2020.1737225] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Revised: 02/24/2020] [Accepted: 02/25/2020] [Indexed: 12/13/2022] Open
Abstract
In utero electroporation (IUE) is a useful technique for gene delivery in embryonic mouse brain. IUE technique is used to investigate the mammalian brain development in vivo. However, according to recent studies, IUE methodology has some limitations like the formation of artificial ectopias and heterotopias at the micro-injection site. Thus far, the artificial heterotopias generated by physical trauma during IUE are rarely reported. Here, we reported the artificial heterotopias and ectopias generated from surgical damages of micropipette in detail, and moreover, we described the protocol to avoid these phenotypes. For the experimental purpose, we transferred empty plasmids (pCAGIG-GFP) with green fluorescent-labelled protein into the cortical cortex by IUE and then compared the structure of the cortex region between the injected and un-injected cerebral hemispheres. The coronary section showed that ectopias and heterotopias were appeared on imperfect-injected brains, and layer maker staining, which including Ctip2 and TBR1 and laminin, can differentiate the physical damage, revealing the neurons in artificial ectopic and heterotopic area were not properly arranged. Moreover, premature differentiation of neurons in ectopias and heterotopias were observed. To avoid heterotopias and ectopias, we carefully manipulated the method of IUE application. Thus, this study might be helpful for the in utero electroporator to distinguish the artificial ectopias and heterotopias that caused by the physical injury by microneedle and the ways to avoid those undesirable circumstances.
Collapse
Affiliation(s)
- Bolin Wang
- School of Pharmacy, Zhejiang Chinese Medical University, Hangzhou, People's Republic of China
| | - Liting Ji
- School of Pharmacy, Zhejiang Chinese Medical University, Hangzhou, People's Republic of China
| | - Kausik Bishayee
- Department of Pharmacology, College of Medicine, Institute of Natural Medicine, Hallym University,Chuncheon, South Korea
| | - Changyu Li
- School of Pharmacy, Zhejiang Chinese Medical University, Hangzhou, People's Republic of China
| | - Sung-Oh Huh
- Department of Pharmacology, College of Medicine, Institute of Natural Medicine, Hallym University,Chuncheon, South Korea
| |
Collapse
|
18
|
Saito T. A Nucleolar Protein, Nepro, Is Essential for the Maintenance of Early Neural Stem Cells and Preimplantation Embryos. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1218:93-101. [PMID: 32060873 DOI: 10.1007/978-3-030-34436-8_6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Notch signaling is required for maintaining neural stem cells (NSCs) in the developing brain. NSCs have potential to give rise to many neuronal types in the early telencephalon, and the potential decreases as embryonic development proceeds. Nepro, which encodes a unique nucleolar protein and is activated downstream of Notch, is essential for maintaining NSCs in the early telencephalon. Nepro is also expressed at basal levels and required for maintaining the preimplantation embryo, by repressing mitochondria-associated p53 apoptotic signaling. Notch signaling also controls dendritic complexity in mitral cells, major projection neurons in the olfactory bulb, showing that many steps of neural development involve Notch signaling.
Collapse
Affiliation(s)
- Tetsuichiro Saito
- Department of Developmental Biology, Graduate School of Medicine, Chiba University, Chiba, Japan.
| |
Collapse
|
19
|
Fujita I, Shitamukai A, Kusumoto F, Mase S, Suetsugu T, Omori A, Kato K, Abe T, Shioi G, Konno D, Matsuzaki F. Endfoot regeneration restricts radial glial state and prevents translocation into the outer subventricular zone in early mammalian brain development. Nat Cell Biol 2019; 22:26-37. [PMID: 31871317 DOI: 10.1038/s41556-019-0436-9] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Accepted: 11/15/2019] [Indexed: 01/19/2023]
Abstract
Neural stem cells, called radial glia, maintain epithelial structure during the early neocortical development. The prevailing view claims that when radial glia first proliferate, their symmetric divisions require strict spindle orientation; its perturbation causes precocious neurogenesis and apoptosis. Here, we show that despite this conventional view, radial glia at the proliferative stage undergo normal symmetric divisions by regenerating an apical endfoot even if it is lost by oblique divisions. We found that the Notch-R-Ras-integrin β1 pathway promotes the regeneration of endfeet, whose leading edge bears ectopic adherens junctions and the Par-polarity complex. However, this regeneration ability gradually declines during the subsequent neurogenic stage and hence oblique divisions induce basal translocation of radial glia to form the outer subventricular zone, a hallmark of the development of the convoluted brain. Our study reveals that endfoot regeneration is a temporally changing cryptic property, which controls the radial glial state and its shift is essential for mammalian brain size expansion.
Collapse
Affiliation(s)
- Ikumi Fujita
- Laboratory for Cell Asymmetry, RIKEN Center for Biosystems Dynamics Research, Kobe, Japan
| | - Atsunori Shitamukai
- Laboratory for Cell Asymmetry, RIKEN Center for Biosystems Dynamics Research, Kobe, Japan
| | - Fumiya Kusumoto
- Laboratory for Cell Asymmetry, RIKEN Center for Biosystems Dynamics Research, Kobe, Japan.,Laboratory of Molecular Cell Biology and Development, Department of Animal Development and Physiology, Graduate School of Biostudies, Kyoto University, Kyoto, Japan
| | - Shun Mase
- Laboratory for Cell Asymmetry, RIKEN Center for Biosystems Dynamics Research, Kobe, Japan.,Laboratory of Molecular Cell Biology and Development, Department of Animal Development and Physiology, Graduate School of Biostudies, Kyoto University, Kyoto, Japan
| | - Taeko Suetsugu
- Laboratory for Cell Asymmetry, RIKEN Center for Biosystems Dynamics Research, Kobe, Japan
| | - Ayaka Omori
- Laboratory for Cell Asymmetry, RIKEN Center for Biosystems Dynamics Research, Kobe, Japan
| | - Kagayaki Kato
- Exploratory Research Center on Life and Living Systems (ExCELLS), National Institutes of Natural Sciences, Tokyo, Japan
| | - Takaya Abe
- Laboratory for Animal Resource Development, RIKEN Center for Biosystems Dynamics Research, Kobe, Japan.,Laboratory of Genetic Engineering, RIKEN Center for Biosystems Dynamics Research, Kobe, Japan
| | - Go Shioi
- Laboratory of Genetic Engineering, RIKEN Center for Biosystems Dynamics Research, Kobe, Japan
| | - Daijiro Konno
- Laboratory for Cell Asymmetry, RIKEN Center for Biosystems Dynamics Research, Kobe, Japan.,Division of Pathophysiology, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan
| | - Fumio Matsuzaki
- Laboratory for Cell Asymmetry, RIKEN Center for Biosystems Dynamics Research, Kobe, Japan. .,Laboratory of Molecular Cell Biology and Development, Department of Animal Development and Physiology, Graduate School of Biostudies, Kyoto University, Kyoto, Japan.
| |
Collapse
|
20
|
Komabayashi-Suzuki M, Yamanishi E, Watanabe C, Okamura M, Tabata H, Iwai R, Ajioka I, Matsushita J, Kidoya H, Takakura N, Okamoto T, Kinoshita K, Ichihashi M, Nagata KI, Ema M, Mizutani KI. Spatiotemporally Dependent Vascularization Is Differently Utilized among Neural Progenitor Subtypes during Neocortical Development. Cell Rep 2019; 29:1113-1129.e5. [DOI: 10.1016/j.celrep.2019.09.048] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2018] [Revised: 06/19/2019] [Accepted: 09/18/2019] [Indexed: 01/07/2023] Open
|
21
|
Suzuki IK. Molecular drivers of human cerebral cortical evolution. Neurosci Res 2019; 151:1-14. [PMID: 31175883 DOI: 10.1016/j.neures.2019.05.007] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2019] [Revised: 05/30/2019] [Accepted: 05/31/2019] [Indexed: 01/10/2023]
Abstract
One of the most important questions in human evolutionary biology is how our ancestor has acquired an expanded volume of the cerebral cortex, which may have significantly impacted on improving our cognitive abilities. Recent comparative approaches have identified developmental features unique to the human or hominid cerebral cortex, not shared with other animals including conventional experimental models. In addition, genomic, transcriptomic, and epigenomic signatures associated with human- or hominid-specific processes of the cortical development are becoming identified by virtue of technical progress in the deep nucleotide sequencing. This review discusses ontogenic and phylogenetic processes of the human cerebral cortex, followed by the introduction of recent comprehensive approaches identifying molecular mechanisms potentially driving the evolutionary changes in the cortical development.
Collapse
Affiliation(s)
- Ikuo K Suzuki
- Department of Biological Sciences, Graduate School of Science, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033 Japan; VIB-KU Leuven Center for Brain & Disease Research, 3000 Leuven, Belgium; Department of Neurosciences, Leuven Brain Institute, KULeuven, 3000 Leuven, Belgium; Université Libre de Bruxelles (U.L.B.), Institut de Recherches en Biologie Humaine et Moléculaire (IRIBHM), ULB Neuroscience Institute (UNI), 1070 Brussels, Belgium.
| |
Collapse
|
22
|
Hirata T, Iwai L. Timing matters: A strategy for neurons to make diverse connections. Neurosci Res 2018; 138:79-83. [PMID: 30227163 DOI: 10.1016/j.neures.2018.09.006] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2018] [Revised: 08/21/2018] [Accepted: 08/21/2018] [Indexed: 11/18/2022]
Abstract
Neurogenesis proceeds like a continuous wave, in which each type of neurons is produced over a few days to several days. During this protracted time window, early-born and late-born neurons are sequentially produced with a considerable time lag. Even if they are identical in their genetic and molecular specifications, they could develop different characteristics under the influences of the timing of their birth. In this review, we discuss the potential influences of "timing" as a generic parameter affecting neuronal differentiation, particularly on axon guidance and connections. These ideas have rarely been tested experimentally, but may provide a new strategy by which phenotypic diversity is increased in neurons.
Collapse
Affiliation(s)
- Tatsumi Hirata
- Division of Brain Function, National Institute of Genetics, 1111 Yata, Mishima, 411-8540, Japan; SOKENDAI (Graduate University for Advanced Studies), Japan.
| | - Lena Iwai
- Division of Brain Function, National Institute of Genetics, 1111 Yata, Mishima, 411-8540, Japan
| |
Collapse
|
23
|
Mizutani KI. Physiological significance of multipolar cells generated from neural stem cells and progenitors for the establishment of neocortical cytoarchitecture. Genes Cells 2017; 23:6-15. [PMID: 29193520 DOI: 10.1111/gtc.12546] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2017] [Accepted: 10/29/2017] [Indexed: 11/28/2022]
Abstract
Neurogenesis encompasses an entire set of events that leads to the generation of newborn neurons from neural stem cells and more committed progenitor cells, including cell division, the production of migratory precursors and their progeny, differentiation and integration into circuits. In particular, the precise control of neuronal migration and morphological changes is essential for the development of the neocortex. Postmitotic cells within the intermediate zone have been found to transiently assume a characteristic "multipolar" morphology, after which a multipolar-to-bipolar transition occurs before the cells enter the cortical plate; however, the importance of this multipolar phase in the establishment of mature cortical cytoarchitecture and the precise genetic control of this phase remains largely unknown. Thus, this review article focuses on the multipolar phase in the developing neocortex. It begins by summarizing the molecular mechanism that underlies multipolar migration for the regulation of each step in multipolar phase in intermediate zone. The physiological significance of this multipolar phase in the establishment of mature cortical lamination and neurodevelopmental disorders associated with migration defects is then described.
Collapse
Affiliation(s)
- Ken-Ichi Mizutani
- Laboratory of Stem Cell Biology, Graduate School of Pharmaceutical Sciences, Kobe Gakuin University, Kobe, Japan
| |
Collapse
|
24
|
A Prdm8 target gene Ebf3 regulates multipolar-to-bipolar transition in migrating neocortical cells. Biochem Biophys Res Commun 2017; 495:388-394. [PMID: 29113800 DOI: 10.1016/j.bbrc.2017.11.021] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2017] [Accepted: 11/03/2017] [Indexed: 01/08/2023]
Abstract
Precise control of neuronal migration is essential for the development of the neocortex. However, the molecular mechanisms underlying neuronal migration remain largely unknown. Here we identified helix-loop-helix transcription factor Ebf3 as a Prdm8 target gene, and found that Ebf3 is a key regulator of neuronal migration via multipolar-to-bipolar transition. Ebf3 knockdown cells exhibited severe defects in the formation of leading processes and an inhibited shift to the locomotion mode. Moreover, we found that Ebf3 knockdown represses NeuroD1 transcription, and NeuroD1 overexpression partially rescued migration defects in Ebf3 knockdown cells. Our findings highlight the critical role of Ebf3 in multipolar-to-bipolar transition via positive feedback regulation of NeuroD1 in the developing neocortex.
Collapse
|
25
|
Lee ML, Martinez-Lozada Z, Krizman EN, Robinson MB. Brain endothelial cells induce astrocytic expression of the glutamate transporter GLT-1 by a Notch-dependent mechanism. J Neurochem 2017; 143:489-506. [PMID: 28771710 DOI: 10.1111/jnc.14135] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2016] [Revised: 07/07/2017] [Accepted: 07/13/2017] [Indexed: 01/13/2023]
Abstract
Neuron-secreted factors induce astrocytic expression of the glutamate transporter, GLT-1 (excitatory amino acid transporter 2). In addition to their elaborate anatomic relationships with neurons, astrocytes also have processes that extend to and envelop the vasculature. Although previous studies have demonstrated that brain endothelia contribute to astrocyte differentiation and maturation, the effects of brain endothelia on astrocytic expression of GLT-1 have not been examined. In this study, we tested the hypothesis that endothelia induce expression of GLT-1 by co-culturing astrocytes from mice that utilize non-coding elements of the GLT-1 gene to control expression of reporter proteins with the mouse endothelial cell line, bEND.3. We found that endothelia increased steady state levels of reporter and GLT-1 mRNA/protein. Co-culturing with primary rat brain endothelia also increases reporter protein, GLT-1 protein, and GLT-1-mediated glutamate uptake. The Janus kinase/signal transducer and activator of transcription 3, bone morphogenic protein/transforming growth factor β, and nitric oxide pathways have been implicated in endothelia-to-astrocyte signaling; we provide multiple lines of evidence that none of these pathways mediate the effects of endothelia on astrocytic GLT-1 expression. Using transwells with a semi-permeable membrane, we demonstrate that the effects of the bEND.3 cell line are dependent upon contact. Notch has also been implicated in endothelia-astrocyte signaling in vitro and in vivo. The first step of Notch signaling requires cleavage of Notch intracellular domain by γ-secretase. We demonstrate that the γ-secretase inhibitor N-[N-(3,5-difluorophenacetyl)-l-alanyl]-S-phenylglycine t-butyl ester blocks endothelia-induced increases in GLT-1. We show that the levels of Notch intracellular domain are higher in nuclei of astrocytes co-cultured with endothelia, an effect also blocked by N-[N-(3,5-difluorophenacetyl)-l-alanyl]-S-phenylglycine t-butyl ester. Finally, infection of co-cultures with shRNA directed against recombination signal binding protein for immunoglobulin kappa J, a Notch effector, also reduces endothelia-dependent increases in enhanced green fluorescent protein and GLT-1. Together, these studies support a novel role for Notch in endothelia-dependent induction of GLT-1 expression. Cover Image for this issue: doi. 10.1111/jnc.13825.
Collapse
Affiliation(s)
- Meredith L Lee
- Departments of Pediatrics, Children's Hospital of Philadelphia, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Zila Martinez-Lozada
- Departments of Pediatrics, Children's Hospital of Philadelphia, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Elizabeth N Krizman
- Departments of Pediatrics, Children's Hospital of Philadelphia, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Michael B Robinson
- Departments of Pediatrics, Children's Hospital of Philadelphia, University of Pennsylvania, Philadelphia, Pennsylvania, USA.,Systems Pharmacology and Translational Therapeutics, Children's Hospital of Philadelphia, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
26
|
Lee ML, Martinez-Lozada Z, Krizman EN, Robinson MB. Brain endothelial cells induce astrocytic expression of the glutamate transporter GLT-1 by a Notch-dependent mechanism. J Neurochem 2017. [PMID: 28771710 DOI: 10.1111/jnc.13825] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Neuron-secreted factors induce astrocytic expression of the glutamate transporter, GLT-1 (excitatory amino acid transporter 2). In addition to their elaborate anatomic relationships with neurons, astrocytes also have processes that extend to and envelop the vasculature. Although previous studies have demonstrated that brain endothelia contribute to astrocyte differentiation and maturation, the effects of brain endothelia on astrocytic expression of GLT-1 have not been examined. In this study, we tested the hypothesis that endothelia induce expression of GLT-1 by co-culturing astrocytes from mice that utilize non-coding elements of the GLT-1 gene to control expression of reporter proteins with the mouse endothelial cell line, bEND.3. We found that endothelia increased steady state levels of reporter and GLT-1 mRNA/protein. Co-culturing with primary rat brain endothelia also increases reporter protein, GLT-1 protein, and GLT-1-mediated glutamate uptake. The Janus kinase/signal transducer and activator of transcription 3, bone morphogenic protein/transforming growth factor β, and nitric oxide pathways have been implicated in endothelia-to-astrocyte signaling; we provide multiple lines of evidence that none of these pathways mediate the effects of endothelia on astrocytic GLT-1 expression. Using transwells with a semi-permeable membrane, we demonstrate that the effects of the bEND.3 cell line are dependent upon contact. Notch has also been implicated in endothelia-astrocyte signaling in vitro and in vivo. The first step of Notch signaling requires cleavage of Notch intracellular domain by γ-secretase. We demonstrate that the γ-secretase inhibitor N-[N-(3,5-difluorophenacetyl)-l-alanyl]-S-phenylglycine t-butyl ester blocks endothelia-induced increases in GLT-1. We show that the levels of Notch intracellular domain are higher in nuclei of astrocytes co-cultured with endothelia, an effect also blocked by N-[N-(3,5-difluorophenacetyl)-l-alanyl]-S-phenylglycine t-butyl ester. Finally, infection of co-cultures with shRNA directed against recombination signal binding protein for immunoglobulin kappa J, a Notch effector, also reduces endothelia-dependent increases in enhanced green fluorescent protein and GLT-1. Together, these studies support a novel role for Notch in endothelia-dependent induction of GLT-1 expression. Cover Image for this issue: doi. 10.1111/jnc.13825.
Collapse
Affiliation(s)
- Meredith L Lee
- Departments of Pediatrics, Children's Hospital of Philadelphia, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Zila Martinez-Lozada
- Departments of Pediatrics, Children's Hospital of Philadelphia, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Elizabeth N Krizman
- Departments of Pediatrics, Children's Hospital of Philadelphia, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Michael B Robinson
- Departments of Pediatrics, Children's Hospital of Philadelphia, University of Pennsylvania, Philadelphia, Pennsylvania, USA.,Systems Pharmacology and Translational Therapeutics, Children's Hospital of Philadelphia, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
27
|
HIV Tat Impairs Neurogenesis through Functioning As a Notch Ligand and Activation of Notch Signaling Pathway. J Neurosci 2017; 36:11362-11373. [PMID: 27807176 DOI: 10.1523/jneurosci.1208-16.2016] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2016] [Accepted: 09/10/2016] [Indexed: 02/07/2023] Open
Abstract
Alterations in adult neurogenesis have been noted in the brain of HIV-infected individuals and are likely linked to HIV-associated neurocognitive deficits, including those in learning and memory. But the underlying molecular mechanisms are not fully understood. In the study, we took advantage of doxycycline-inducible and astrocyte-specific HIV-1 Tat transgenic mice (iTat) and determined the relationship between Tat expression and neurogenesis. Tat expression in astrocytes was associated with fewer neuron progenitor cells (NPCs), fewer immature neurons, and fewer mature neurons in the dentate gyrus of the hippocampus of the mouse brain. In vitro NPC-derived neurosphere assays showed that Tat-containing conditioned media from astrocytes or recombinant Tat protein inhibited NPC proliferation and migration and altered NPC differentiation, while immunodepletion of Tat from Tat-containing conditioned media or heat inactivation of recombinant Tat abrogated those effects. Notch signaling downstream gene Hes1 promoter-driven luciferase reporter gene assay and Western blotting showed that recombinant Tat or Tat-containing conditioned media activated Hes1 transcription and protein expression, which were abrogated by Tat heat inactivation, immunodepletion, and cysteine mutation at position 30. Last, Notch signaling inhibitor N-[N-(3,5-difluorophenacetyl)-l-alanyl]-S-phenylglycine t-butyl ester (DAPT) significantly rescued Tat-impaired NPC differentiation in vitro and neurogenesis in vivo Together, these results show that Tat adversely affects NPCs and neurogenesis through Notch signaling and point to the potential of developing Notch signaling inhibitors as HIV/neuroAIDS therapeutics. SIGNIFICANCE STATEMENT HIV infection of the CNS causes cognitive and memory deficits, which have become more prevalent in the era of combination antiretroviral therapy (cART). Neurogenesis is impaired in HIV-infected individuals. But the underlying molecular mechanisms remain largely unknown. In this study, we have discovered that HIV Tat impairs neurogenesis through the Notch signaling pathway. These findings are particularly important because Tat protein has recently been detected in the brain of HIV-infected individuals with HIV replication in the periphery being effectively controlled by cART. The current study not only further highlights the importance of HIV Tat protein in HIV/neuroAIDS, but also presents a new strategy to develop novel HIV/neuroAIDS therapeutics, particularly in the era of cART.
Collapse
|
28
|
Roese-Koerner B, Stappert L, Brüstle O. Notch/Hes signaling and miR-9 engage in complex feedback interactions controlling neural progenitor cell proliferation and differentiation. NEUROGENESIS 2017; 4:e1313647. [PMID: 28573150 PMCID: PMC5443189 DOI: 10.1080/23262133.2017.1313647] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/01/2016] [Revised: 11/27/2016] [Accepted: 01/18/2017] [Indexed: 02/04/2023]
Abstract
Canonical Notch signaling has diverse functions during nervous system development and is critical for neural progenitor self-renewal, timing of differentiation and specification of various cell fates. A key feature of Notch-mediated self-renewal is its fluctuating activity within the neural progenitor cell population and the oscillatory expression pattern of the Notch effector Hes1 and its target genes. A negative feedback loop between Hes1 and neurogenic microRNA miR-9 was found to be part of this oscillatory clock. In a recent study we discovered that miR-9 expression is further modulated by direct binding of the Notch intracellular domain/RBPj transcriptional complex to the miR-9_2 promoter. In turn, miR-9 not only targets Hes1 but also Notch2 to attenuate Notch signaling and promote neuronal differentiation. Here, we discuss how the two interwoven feedback loops may provide an additional fail-save mechanism to control proliferation and differentiation within the neural progenitor cell population. Furthermore, we explore potential implications of miR-9-mediated regulation of Notch/Hes1 signaling with regard to neural progenitor homeostasis, patterning, timing of differentiation and tumor formation.
Collapse
Affiliation(s)
- Beate Roese-Koerner
- Institute of Reconstructive Neurobiology, LIFE & BRAIN Center, University of Bonn Medical Faculty, Bonn, Germany
| | - Laura Stappert
- Institute of Reconstructive Neurobiology, LIFE & BRAIN Center, University of Bonn Medical Faculty, Bonn, Germany
| | - Oliver Brüstle
- Institute of Reconstructive Neurobiology, LIFE & BRAIN Center, University of Bonn Medical Faculty, Bonn, Germany
| |
Collapse
|
29
|
Muroyama Y, Baba A, Kitagawa M, Saito T. Olfactory Sensory Neurons Control Dendritic Complexity of Mitral Cells via Notch Signaling. PLoS Genet 2016; 12:e1006514. [PMID: 28027303 PMCID: PMC5189955 DOI: 10.1371/journal.pgen.1006514] [Citation(s) in RCA: 11] [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: 08/03/2016] [Accepted: 11/29/2016] [Indexed: 11/29/2022] Open
Abstract
Mitral cells (MCs) of the mammalian olfactory bulb have a single primary dendrite extending into a single glomerulus, where they receive odor information from olfactory sensory neurons (OSNs). Molecular mechanisms for controlling dendritic arbors of MCs, which dynamically change during development, are largely unknown. Here we found that MCs displayed more complex dendritic morphologies in mouse mutants of Maml1, a crucial gene in Notch signaling. Similar phenotypes were observed by conditionally misexpressing a dominant negative form of MAML1 (dnMAML1) in MCs after their migration. Conversely, conditional misexpression of a constitutively active form of Notch reduced their dendritic complexity. Furthermore, the intracellular domain of Notch1 (NICD1) was localized to nuclei of MCs. These findings suggest that Notch signaling at embryonic stages is involved in the dendritic complexity of MCs. After the embryonic misexpression of dnMAML1, many MCs aberrantly extended dendrites to more than one glomerulus at postnatal stages, suggesting that Notch signaling is essential for proper formation of olfactory circuits. Moreover, dendrites in cultured MCs were shortened by Jag1-expressing cells. Finally, blocking the activity of Notch ligands in OSNs led to an increase in dendritic complexity as well as a decrease in NICD1 signals in MCs. These results demonstrate that the dendritic complexity of MCs is controlled by their presynaptic partners, OSNs. Olfactory circuits are critical for the survival of many animals. Odor information is transmitted from olfactory sensory neurons (OSNs) to relay neurons, the morphology of which is crucial for processing of the information and similar among species. The major relay neurons, mitral cells (MCs) in mammals and projection neurons in flies, have a single primary dendrite at the mature stage. Molecular mechanisms to control the formation of the dendrite are largely unknown. MCs dynamically change their dendrites during development. In this study, we show that the dendritic morphologies of MCs are controlled by Notch signaling, many factors of which are well conserved among species. Moreover, we have found that Notch signaling in MCs is activated by OSNs, and that Notch operates in the relay neurons in the mouse olfactory system, in contrast to the fly system, where Notch functions in OSNs. Therefore, our study has revealed a novel step for shaping the dendritic morphologies of MCs.
Collapse
Affiliation(s)
- Yuko Muroyama
- Department of Developmental Biology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Atsushi Baba
- Department of Developmental Biology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Motoo Kitagawa
- Department of Molecular and Tumor Pathology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Tetsuichiro Saito
- Department of Developmental Biology, Graduate School of Medicine, Chiba University, Chiba, Japan
- * E-mail:
| |
Collapse
|
30
|
Inoue M, Iwai R, Tabata H, Konno D, Komabayashi-Suzuki M, Watanabe C, Iwanari H, Mochizuki Y, Hamakubo T, Matsuzaki F, Nagata KI, Mizutani KI. Prdm16 is crucial for progression of the multipolar phase during neural differentiation of the developing neocortex. Development 2016; 144:385-399. [PMID: 27993981 DOI: 10.1242/dev.136382] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2016] [Accepted: 12/06/2016] [Indexed: 01/09/2023]
Abstract
The precise control of neuronal migration and morphological changes during differentiation is essential for neocortical development. We hypothesized that the transition of progenitors through progressive stages of differentiation involves dynamic changes in levels of mitochondrial reactive oxygen species (mtROS), depending on cell requirements. We found that progenitors had higher levels of mtROS, but that these levels were significantly decreased with differentiation. The Prdm16 gene was identified as a candidate modulator of mtROS using microarray analysis, and was specifically expressed by progenitors in the ventricular zone. However, Prdm16 expression declined during the transition into NeuroD1-positive multipolar cells. Subsequently, repression of Prdm16 expression by NeuroD1 on the periphery of ventricular zone was crucial for appropriate progression of the multipolar phase and was required for normal cellular development. Furthermore, time-lapse imaging experiments revealed abnormal migration and morphological changes in Prdm16-overexpressing and -knockdown cells. Reporter assays and mtROS determinations demonstrated that PGC1α is a major downstream effector of Prdm16 and NeuroD1, and is required for regulation of the multipolar phase and characteristic modes of migration. Taken together, these data suggest that Prdm16 plays an important role in dynamic cellular redox changes in developing neocortex during neural differentiation.
Collapse
Affiliation(s)
- Mayuko Inoue
- Laboratory of Neural Differentiation, Graduate School of Brain Science, Doshisha University, Kyoto 6190225, Japan
| | - Ryota Iwai
- Laboratory of Neural Differentiation, Graduate School of Brain Science, Doshisha University, Kyoto 6190225, Japan
| | - Hidenori Tabata
- Department of Molecular Neurobiology, Institute for Developmental Research, Kasugai, Aichi 4800392, Japan
| | - Daijiro Konno
- Laboratory for Cell Asymmetry, Center for Developmental Biology, RIKEN Kobe Institute, 2-2-3 Minatojima-minamimachi, Chuo-ku, Kobe, Hyogo 650-0047, Japan
| | - Mariko Komabayashi-Suzuki
- Laboratory of Neural Differentiation, Graduate School of Brain Science, Doshisha University, Kyoto 6190225, Japan
| | - Chisato Watanabe
- Laboratory of Neural Differentiation, Graduate School of Brain Science, Doshisha University, Kyoto 6190225, Japan
| | - Hiroko Iwanari
- Department of Quantitative Biology and Medicine, RCAST, The University of Tokyo, Tokyo 1538904, Japan
| | - Yasuhiro Mochizuki
- Department of Quantitative Biology and Medicine, RCAST, The University of Tokyo, Tokyo 1538904, Japan
| | - Takao Hamakubo
- Department of Quantitative Biology and Medicine, RCAST, The University of Tokyo, Tokyo 1538904, Japan
| | - Fumio Matsuzaki
- Laboratory for Cell Asymmetry, Center for Developmental Biology, RIKEN Kobe Institute, 2-2-3 Minatojima-minamimachi, Chuo-ku, Kobe, Hyogo 650-0047, Japan
| | - Koh-Ichi Nagata
- Department of Molecular Neurobiology, Institute for Developmental Research, Kasugai, Aichi 4800392, Japan
| | - Ken-Ichi Mizutani
- Laboratory of Neural Differentiation, Graduate School of Brain Science, Doshisha University, Kyoto 6190225, Japan .,PRESTO "Development and Function of Neural Networks", Japan Science and Technology Agency, Saitama 3320012, Japan
| |
Collapse
|
31
|
Suzuki K, Tsunekawa Y, Hernandez-Benitez R, Wu J, Zhu J, Kim EJ, Hatanaka F, Yamamoto M, Araoka T, Li Z, Kurita M, Hishida T, Li M, Aizawa E, Guo S, Chen S, Goebl A, Soligalla RD, Qu J, Jiang T, Fu X, Jafari M, Esteban CR, Berggren WT, Lajara J, Nuñez-Delicado E, Guillen P, Campistol JM, Matsuzaki F, Liu GH, Magistretti P, Zhang K, Callaway EM, Zhang K, Belmonte JCI. In vivo genome editing via CRISPR/Cas9 mediated homology-independent targeted integration. Nature 2016; 540:144-149. [PMID: 27851729 DOI: 10.1038/nature20565] [Citation(s) in RCA: 838] [Impact Index Per Article: 93.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2016] [Accepted: 10/27/2016] [Indexed: 02/05/2023]
Abstract
Targeted genome editing via engineered nucleases is an exciting area of biomedical research and holds potential for clinical applications. Despite rapid advances in the field, in vivo targeted transgene integration is still infeasible because current tools are inefficient, especially for non-dividing cells, which compose most adult tissues. This poses a barrier for uncovering fundamental biological principles and developing treatments for a broad range of genetic disorders. Based on clustered regularly interspaced short palindromic repeat/Cas9 (CRISPR/Cas9) technology, here we devise a homology-independent targeted integration (HITI) strategy, which allows for robust DNA knock-in in both dividing and non-dividing cells in vitro and, more importantly, in vivo (for example, in neurons of postnatal mammals). As a proof of concept of its therapeutic potential, we demonstrate the efficacy of HITI in improving visual function using a rat model of the retinal degeneration condition retinitis pigmentosa. The HITI method presented here establishes new avenues for basic research and targeted gene therapies.
Collapse
Affiliation(s)
- Keiichiro Suzuki
- Gene Expression Laboratory, Salk Institute for Biological Studies, 10010 N. Torrey Pines Rd, La Jolla, California 92037, USA
| | - Yuji Tsunekawa
- Laboratory for Cell Asymmetry, RIKEN Center for Developmental Biology, 2-2-3 Minatojima-Minamimachi, Chuo-ku, Kobe 650-0047, Japan
| | - Reyna Hernandez-Benitez
- Gene Expression Laboratory, Salk Institute for Biological Studies, 10010 N. Torrey Pines Rd, La Jolla, California 92037, USA.,4700 King Abdullah University of Science and Technology (KAUST) Thuwal 23955-6900, Saudi Arabia
| | - Jun Wu
- Gene Expression Laboratory, Salk Institute for Biological Studies, 10010 N. Torrey Pines Rd, La Jolla, California 92037, USA.,Universidad Católica San Antonio de Murcia (UCAM) Campus de los Jerónimos, no. 135 Guadalupe 30107, Murcia, Spain
| | - Jie Zhu
- Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510623, China.,Shiley Eye Institute, Institute for Genomic Medicine, Institute of Engineering in Medicine, University of California, San Diego, 9500 Gilman Drive #0946, La Jolla, California 92023, USA
| | - Euiseok J Kim
- Systems Neurobiology Laboratory, Salk Institute for Biological Studies, 10010 N. Torrey Pines Rd., La Jolla, California 92037, USA
| | - Fumiyuki Hatanaka
- Gene Expression Laboratory, Salk Institute for Biological Studies, 10010 N. Torrey Pines Rd, La Jolla, California 92037, USA
| | - Mako Yamamoto
- Gene Expression Laboratory, Salk Institute for Biological Studies, 10010 N. Torrey Pines Rd, La Jolla, California 92037, USA
| | - Toshikazu Araoka
- Gene Expression Laboratory, Salk Institute for Biological Studies, 10010 N. Torrey Pines Rd, La Jolla, California 92037, USA.,Universidad Católica San Antonio de Murcia (UCAM) Campus de los Jerónimos, no. 135 Guadalupe 30107, Murcia, Spain
| | - Zhe Li
- Bioengineering, University of California, San Diego, 9500 Gilman Drive, MC0412, La Jolla, California 92093-0412, USA
| | - Masakazu Kurita
- Gene Expression Laboratory, Salk Institute for Biological Studies, 10010 N. Torrey Pines Rd, La Jolla, California 92037, USA
| | - Tomoaki Hishida
- Gene Expression Laboratory, Salk Institute for Biological Studies, 10010 N. Torrey Pines Rd, La Jolla, California 92037, USA
| | - Mo Li
- Gene Expression Laboratory, Salk Institute for Biological Studies, 10010 N. Torrey Pines Rd, La Jolla, California 92037, USA
| | - Emi Aizawa
- Gene Expression Laboratory, Salk Institute for Biological Studies, 10010 N. Torrey Pines Rd, La Jolla, California 92037, USA
| | - Shicheng Guo
- Bioengineering, University of California, San Diego, 9500 Gilman Drive, MC0412, La Jolla, California 92093-0412, USA
| | - Song Chen
- Bioengineering, University of California, San Diego, 9500 Gilman Drive, MC0412, La Jolla, California 92093-0412, USA
| | - April Goebl
- Gene Expression Laboratory, Salk Institute for Biological Studies, 10010 N. Torrey Pines Rd, La Jolla, California 92037, USA
| | - Rupa Devi Soligalla
- Gene Expression Laboratory, Salk Institute for Biological Studies, 10010 N. Torrey Pines Rd, La Jolla, California 92037, USA
| | - Jing Qu
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China.,University of Chinese Academy of Sciences, Beijing 100049, China
| | - Tingshuai Jiang
- Shiley Eye Institute, Institute for Genomic Medicine, Institute of Engineering in Medicine, University of California, San Diego, 9500 Gilman Drive #0946, La Jolla, California 92023, USA.,Guangzhou EliteHealth Biological Pharmaceutical Technology Company Ltd, Guangzhou 510005, China
| | - Xin Fu
- Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510623, China.,Shiley Eye Institute, Institute for Genomic Medicine, Institute of Engineering in Medicine, University of California, San Diego, 9500 Gilman Drive #0946, La Jolla, California 92023, USA
| | - Maryam Jafari
- Shiley Eye Institute, Institute for Genomic Medicine, Institute of Engineering in Medicine, University of California, San Diego, 9500 Gilman Drive #0946, La Jolla, California 92023, USA
| | - Concepcion Rodriguez Esteban
- Gene Expression Laboratory, Salk Institute for Biological Studies, 10010 N. Torrey Pines Rd, La Jolla, California 92037, USA
| | - W Travis Berggren
- Salk Institute for Biological Studies, 10010 N. Torrey Pines Rd, La Jolla, California 92037, USA
| | - Jeronimo Lajara
- Universidad Católica San Antonio de Murcia (UCAM) Campus de los Jerónimos, no. 135 Guadalupe 30107, Murcia, Spain
| | - Estrella Nuñez-Delicado
- Universidad Católica San Antonio de Murcia (UCAM) Campus de los Jerónimos, no. 135 Guadalupe 30107, Murcia, Spain
| | - Pedro Guillen
- Universidad Católica San Antonio de Murcia (UCAM) Campus de los Jerónimos, no. 135 Guadalupe 30107, Murcia, Spain.,Fundación Dr. Pedro Guillen, Investigación Biomedica de Clinica CEMTRO, Avenida Ventisquero de la Condesa, 42, 28035 Madrid, Spain
| | - Josep M Campistol
- Hospital Clinic, University of Barcelona, IDIBAPS, 08036 Barcelona, Spain
| | - Fumio Matsuzaki
- Laboratory for Cell Asymmetry, RIKEN Center for Developmental Biology, 2-2-3 Minatojima-Minamimachi, Chuo-ku, Kobe 650-0047, Japan
| | - Guang-Hui Liu
- University of Chinese Academy of Sciences, Beijing 100049, China.,National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China.,Key Laboratory of Regenerative Medicine of Ministry of Education, Institute of Aging and Regenerative Medicine, Jinan University, Guangzhou 510632, China.,Beijing Institute for Brain Disorders, Beijing 100069, China
| | - Pierre Magistretti
- 4700 King Abdullah University of Science and Technology (KAUST) Thuwal 23955-6900, Saudi Arabia
| | - Kun Zhang
- Bioengineering, University of California, San Diego, 9500 Gilman Drive, MC0412, La Jolla, California 92093-0412, USA
| | - Edward M Callaway
- Systems Neurobiology Laboratory, Salk Institute for Biological Studies, 10010 N. Torrey Pines Rd., La Jolla, California 92037, USA
| | - Kang Zhang
- Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510623, China.,Shiley Eye Institute, Institute for Genomic Medicine, Institute of Engineering in Medicine, University of California, San Diego, 9500 Gilman Drive #0946, La Jolla, California 92023, USA.,Molecular Medicine Research Center, West China Hospital, Sichuan University, Chengdu 610041, China.,Veterans Administration Healthcare System, San Diego, California 92093, USA
| | - Juan Carlos Izpisua Belmonte
- Gene Expression Laboratory, Salk Institute for Biological Studies, 10010 N. Torrey Pines Rd, La Jolla, California 92037, USA
| |
Collapse
|
32
|
Kim KT, Song MR. Light-induced Notch activity controls neurogenic and gliogenic potential of neural progenitors. Biochem Biophys Res Commun 2016; 479:820-826. [PMID: 27680314 DOI: 10.1016/j.bbrc.2016.09.124] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2016] [Accepted: 09/24/2016] [Indexed: 11/27/2022]
Abstract
Oscillations in Notch signaling are essential for reserving neural progenitors for cellular diversity in developing brains. Thus, steady and prolonged overactivation of Notch signaling is not suitable for generating neurons. To acquire greater temporal control of Notch activity and mimic endogenous oscillating signals, here we adopted a light-inducible transgene system to induce active form of Notch NICD in neural progenitors. Alternating Notch activity saved more progenitors that are prone to produce neurons creating larger number of mixed clones with neurons and progenitors in vitro, compared to groups with no light or continuous light stimulus. Furthermore, more upper layer neurons and astrocytes arose upon intermittent Notch activity, indicating that dynamic Notch activity maintains neural progeny and fine-tune neuron-glia diversity.
Collapse
Affiliation(s)
- Kyung-Tai Kim
- School of Life Sciences, Gwangju Institute of Science and Technology, Oryong-dong, Buk-gu, Gwangju 500-712, Republic of Korea
| | - Mi-Ryoung Song
- School of Life Sciences, Gwangju Institute of Science and Technology, Oryong-dong, Buk-gu, Gwangju 500-712, Republic of Korea.
| |
Collapse
|
33
|
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
|
34
|
Cell-cycle-independent transitions in temporal identity of mammalian neural progenitor cells. Nat Commun 2016; 7:11349. [PMID: 27094546 PMCID: PMC4842982 DOI: 10.1038/ncomms11349] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2016] [Accepted: 03/17/2016] [Indexed: 12/30/2022] Open
Abstract
During cerebral development, many types of neurons are sequentially generated by self-renewing progenitor cells called apical progenitors (APs). Temporal changes in AP identity are thought to be responsible for neuronal diversity; however, the mechanisms underlying such changes remain largely unknown. Here we perform single-cell transcriptome analysis of individual progenitors at different developmental stages, and identify a subset of genes whose expression changes over time but is independent of differentiation status. Surprisingly, the pattern of changes in the expression of such temporal-axis genes in APs is unaffected by cell-cycle arrest. Consistent with this, transient cell-cycle arrest of APs in vivo does not prevent descendant neurons from acquiring their correct laminar fates. Analysis of cultured APs reveals that transitions in AP gene expression are driven by both cell-intrinsic and -extrinsic mechanisms. These results suggest that the timing mechanisms controlling AP temporal identity function independently of cell-cycle progression and Notch activation mode.
Collapse
|
35
|
Wang W, Jossin Y, Chai G, Lien WH, Tissir F, Goffinet AM. Feedback regulation of apical progenitor fate by immature neurons through Wnt7-Celsr3-Fzd3 signalling. Nat Commun 2016; 7:10936. [PMID: 26939553 PMCID: PMC4786577 DOI: 10.1038/ncomms10936] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2015] [Accepted: 02/03/2016] [Indexed: 11/09/2022] Open
Abstract
Sequential generation of neurons and glial cells during development is critical for the wiring and function of the cerebral cortex. This process requires accurate coordination of neural progenitor cell (NPC) fate decisions, by NPC-autonomous mechanisms as well as by negative feedback from neurons. Here, we show that neurogenesis is protracted and gliogenesis decreased in mice with mutations of genes Celsr3 and Fzd3. This phenotype is not due to gene inactivation in progenitors, but rather in immature cortical neurons. Mutant neurons are unable to upregulate expression of Jag1 in response to cortical Wnt7, resulting in blunted activation of Notch signalling in NPC. Thus, Celsr3 and Fzd3 enable immature neurons to respond to Wnt7, upregulate Jag1 and thereby facilitate feedback signals that tune the timing of NPC fate decisions via Notch activation.
Collapse
Affiliation(s)
- Wei Wang
- Université catholique de Louvain, Institute of Neuroscience, 73 Avenue Mounier, Box B1.7316, 1200 Brussels, Belgium
| | - Yves Jossin
- Université catholique de Louvain, Institute of Neuroscience, 73 Avenue Mounier, Box B1.7316, 1200 Brussels, Belgium
| | - Guoliang Chai
- Université catholique de Louvain, Institute of Neuroscience, 73 Avenue Mounier, Box B1.7316, 1200 Brussels, Belgium
| | - Wen-Hui Lien
- Université catholique de Louvain, de Duve Institute, Avenue Hippocrate 74, Box B1.74.09, 1200 Brussels, Belgium
| | - Fadel Tissir
- Université catholique de Louvain, Institute of Neuroscience, 73 Avenue Mounier, Box B1.7316, 1200 Brussels, Belgium
| | - Andre M Goffinet
- Université catholique de Louvain, Institute of Neuroscience, 73 Avenue Mounier, Box B1.7316, 1200 Brussels, Belgium.,WELBIO, 6 Avenue Pasteur, 1300 Wavre, Belgium
| |
Collapse
|
36
|
Mirra S, Ulloa F, Gutierrez-Vallejo I, Martì E, Soriano E. Function of Armcx3 and Armc10/SVH Genes in the Regulation of Progenitor Proliferation and Neural Differentiation in the Chicken Spinal Cord. Front Cell Neurosci 2016; 10:47. [PMID: 26973462 PMCID: PMC4776218 DOI: 10.3389/fncel.2016.00047] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2015] [Accepted: 02/09/2016] [Indexed: 12/03/2022] Open
Abstract
The eutherian X-chromosome specific family of Armcx genes has been described as originating by retrotransposition from Armc10/SVH, a single Arm-containing somatic gene. Armcx3 and Armc10/SVH are characterized by high expression in the central nervous system and they play an important role in the regulation of mitochondrial distribution and transport in neurons. In addition, Armcx/Arm10 genes have several Armadillo repeats in their sequence. In this study we address the potential role of this gene family in neural development by using the chick neural tube as a model. We show that Armc10/SVH is expressed in the chicken spinal cord, and knocking-down Armc10/SVH by sh-RNAi electroporation in spinal cord reduces proliferation of neural precursor cells (NPCs). Moreover, we analyzed the effects of murine Armcx3 and Armc10 overexpression, showing that both proteins regulate progenitor proliferation, while Armcx3 overexpression also specifically controls neural maturation. We show that the phenotypes found following Armcx3 overexpression require its mitochondrial localization, suggesting a novel link between mitochondrial dynamics and regulation of neural development. Furthermore, we found that both Armcx3 and Armc10 may act as inhibitors of Wnt-β-catenin signaling. Our results highlight both common and differential functions of Armcx/Armc10 genes in neural development in the spinal cord.
Collapse
Affiliation(s)
- Serena Mirra
- Department of Cell Biology, Faculty of Biology, University of BarcelonaBarcelona, Spain; Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas, Instituto de Salud Carlos IIIMadrid, Spain
| | - Fausto Ulloa
- Department of Cell Biology, Faculty of Biology, University of BarcelonaBarcelona, Spain; Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas, Instituto de Salud Carlos IIIMadrid, Spain
| | - Irene Gutierrez-Vallejo
- Instituto de Biología Molecular de Barcelona, Consejo Superior de Investigaciones Científicas, ParcCientífic de Barcelona Barcelona, Spain
| | - Elisa Martì
- Instituto de Biología Molecular de Barcelona, Consejo Superior de Investigaciones Científicas, ParcCientífic de Barcelona Barcelona, Spain
| | - Eduardo Soriano
- Department of Cell Biology, Faculty of Biology, University of BarcelonaBarcelona, Spain; Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas, Instituto de Salud Carlos IIIMadrid, Spain; Valld'Hebron Institute of ResearchBarcelona, Spain; Institució Catalana de Recerca i Estudis AvançatsBarcelona, Spain
| |
Collapse
|
37
|
Baumgart J, Baumgart N. Cortex-, Hippocampus-, Thalamus-, Hypothalamus-, Lateral Septal Nucleus- and Striatum-specific In Utero Electroporation in the C57BL/6 Mouse. J Vis Exp 2016:e53303. [PMID: 26862715 DOI: 10.3791/53303] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022] Open
Abstract
In utero electroporation is a widely used technique for fast and efficient spatiotemporal manipulation of various genes in the rodent central nervous system. Overexpression of desired genes is just as possible as shRNA mediated loss-of-function studies. Therefore it offers a wide range of applications. The feasibility to target particular cells in a distinct area further increases the range of potential applications of this very useful method. For efficiently targeting specific regions knowledge about the subtleties, such as the embryonic stage, the voltage to apply and most importantly the position of the electrodes, is indispensable. Here, we provide a detailed protocol that allows for specific and efficient in utero electroporation of several regions of the C57BL/6 mouse central nervous system. In particular it is shown how to transfect regions the develop into the retrosplenial cortex, the motor cortex, the somatosensory cortex, the piriform cortex, the cornu ammonis 1-3, the dentate gyrus, the striatum, the lateral septal nucleus, the thalamus and the hypothalamus. For this information about the appropriate embryonic stage, the appropriate voltage for the corresponding embryonic stage is provided. Most importantly an angle-map, which indicates the appropriate position of the positive pole, is depicted. This standardized protocol helps to facilitate efficient in utero electroporation, which might also lead to a reduced number of animals.
Collapse
Affiliation(s)
- Jan Baumgart
- TARC (Translational Animal Research Center), University Medical Center, JGU Mainz
| | - Nadine Baumgart
- TARC (Translational Animal Research Center), University Medical Center, JGU Mainz;
| |
Collapse
|
38
|
Inoue M, Iwai R, Yamanishi E, Yamagata K, Komabayashi-Suzuki M, Honda A, Komai T, Miyachi H, Kitano S, Watanabe C, Teshima W, Mizutani KI. Deletion of Prdm8 impairs development of upper-layer neocortical neurons. Genes Cells 2015; 20:758-70. [PMID: 26283595 DOI: 10.1111/gtc.12274] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2015] [Accepted: 06/15/2015] [Indexed: 01/17/2023]
Abstract
Upper-layer (UL) neocortical neurons are the most prominent distinguishing features of the mammalian neocortex compared with those of the avian dorsal cortex and are vastly expanded in primates. However, little is known about the identities of the genes that control the specification of UL neurons. Here, we found that Prdm8, a member of the PR (PRDI-BF1 and RIZ homology) domain protein family, was specifically expressed in the postnatal UL neocortex, particular those in late-born RORß-positive layer IV neurons. We generated homozygous Prdm8 knockout (Prdm8 KO) mice and found that the deletion of Prdm8 causes growth retardation and a reduced brain weight, although the brain weight-to-body weight ratio is unchanged at postnatal day 8 (P8). Immunohistochemistry showed that the relative UL thickness, but not the thickness of the deep layer (DL), was significantly reduced in Prdm8 KO mice compared with wild-type (WT) mice. In addition, we found that a number of late-born Brn2-positive UL neurons were significantly decreased in Prdm8 KO mice. To identify genes regulated by Prdm8 during neocortical development, we compared expression profiling analysis in Prdm8 KO and WT mice, and identified some candidate genes. These results suggest that the proper expression of Prdm8 is required for the normal development and construction of UL neurons in the mammalian neocortex.
Collapse
Affiliation(s)
- Mayuko Inoue
- Laboratory of Neural Differentiation, Graduate School of Brain Science, Doshisha University, Kyoto, 619-0225, Japan.,Institute for Virus Research, Kyoto University, Kyoto, 606-8507, Japan
| | - Ryota Iwai
- Laboratory of Neural Differentiation, Graduate School of Brain Science, Doshisha University, Kyoto, 619-0225, Japan
| | - Emiko Yamanishi
- Laboratory of Neural Differentiation, Graduate School of Brain Science, Doshisha University, Kyoto, 619-0225, Japan
| | - Kazuyuki Yamagata
- Laboratory of Neural Differentiation, Graduate School of Brain Science, Doshisha University, Kyoto, 619-0225, Japan
| | - Mariko Komabayashi-Suzuki
- Laboratory of Neural Differentiation, Graduate School of Brain Science, Doshisha University, Kyoto, 619-0225, Japan
| | - Aya Honda
- Laboratory of Neural Differentiation, Graduate School of Brain Science, Doshisha University, Kyoto, 619-0225, Japan
| | - Tae Komai
- Institute for Virus Research, Kyoto University, Kyoto, 606-8507, Japan
| | - Hitoshi Miyachi
- Institute for Virus Research, Kyoto University, Kyoto, 606-8507, Japan
| | - Satsuki Kitano
- Institute for Virus Research, Kyoto University, Kyoto, 606-8507, Japan
| | - Chisato Watanabe
- Laboratory of Neural Differentiation, Graduate School of Brain Science, Doshisha University, Kyoto, 619-0225, Japan
| | - Waka Teshima
- Laboratory of Neural Differentiation, Graduate School of Brain Science, Doshisha University, Kyoto, 619-0225, Japan
| | - Ken-ichi Mizutani
- Laboratory of Neural Differentiation, Graduate School of Brain Science, Doshisha University, Kyoto, 619-0225, Japan.,Japan Science and Technology Agency, PRESTO, Saitama, 332-0012, Japan
| |
Collapse
|
39
|
Yamanishi E, Yoon K, Alberi L, Gaiano N, Mizutani KI. NF-κB signaling regulates the generation of intermediate progenitors in the developing neocortex. Genes Cells 2015; 20:706-19. [PMID: 26243725 DOI: 10.1111/gtc.12267] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2015] [Accepted: 05/17/2015] [Indexed: 01/22/2023]
Abstract
In addition to its well-established role during immune system function, NF-κB regulates cell survival and synaptic plasticity in the mature nervous system. Here, we show that during mouse brain development, NF-κB activity is present in the neocortical ventricular and subventricular zones (VZ and SVZ), where it regulates proliferative pool maintenance. Activation of NF-κB signaling, by expression of p65 or an activated form of the IκB kinase complex subunit IKK2, inhibited neuronal differentiation and promoted retention of progenitors in the VZ and SVZ. In contrast, blockade of the pathway with dominant negative forms of IKK2 and IκBα promoted neuronal differentiation both in vivo and in vitro. Furthermore, by modulating both the NF-κB and Notch pathways, we show that in the absence of canonical Notch activity, after knockdown of the pathway effector CBF1, NF-κB signaling promoted Tbr2 expression and intermediate neural progenitor fate. Interestingly, however, activation of NF-κB in vivo, with canonical Notch signaling intact, promoted expression of the radial glial marker Pax6. This work identifies NF-κB signaling as a regulator of neocortical neurogenesis and suggests that the pathway plays roles in both the VZ and SVZ.
Collapse
Affiliation(s)
- Emiko Yamanishi
- Laboratory of Neural Differentiation, Graduate School of Brain Science, Doshisha University, 1-3 Tatara Miyakodani, Kyotanabe-shi, Kyoto, 610-0394, Japan
| | - Keejung Yoon
- Neuroregeneration Program, Institute for Cell Engineering, Departments of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Lavinia Alberi
- Neuroregeneration Program, Institute for Cell Engineering, Departments of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Nicholas Gaiano
- Neuroregeneration Program, Institute for Cell Engineering, Departments of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Ken-ichi Mizutani
- Laboratory of Neural Differentiation, Graduate School of Brain Science, Doshisha University, 1-3 Tatara Miyakodani, Kyotanabe-shi, Kyoto, 610-0394, Japan.,Neuroregeneration Program, Institute for Cell Engineering, Departments of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA.,Japan Science and Technology Agency, PRESTO, Saitama, 332-0012, Japan
| |
Collapse
|
40
|
|
41
|
C57BL/6-specific conditions for efficient in utero electroporation of the central nervous system. J Neurosci Methods 2014; 240:116-24. [PMID: 25445056 DOI: 10.1016/j.jneumeth.2014.11.004] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2014] [Revised: 09/27/2014] [Accepted: 11/05/2014] [Indexed: 01/28/2023]
Abstract
BACKGROUND In utero electroporation is a fast an efficient tool to specifically address gene expression in the murine central nervous system. This technique was originally established in ICR/CD-1 outbred mice. Neuroanatomical differences between the different mouse strains and variations in gestation length require the optimization of the conditions for each strain to avoid severe complications. Furthermore the relevant position information is currently only scarcely standardized and not always easy to transfer to C57BL/6 mice. NEW METHOD In this study we present an improved method for in utero electroporation of C57BL/6 including a detailed atlas that allows for specific and efficient in vivo transfection. Further we introduce histogram analysis as a tool for neural migration assays. RESULTS We report individually adapted conditions for in utero electroporation in C57BL/6 mice that differ from the previously published data for ICR/CD-1 mice. Furthermore, this article outlines a detailed angle-map that allows for the specific and efficient in vivo transfection of different regions of the C57BL/6 mouse central nervous system. We also show that histogram analysis is a valuable tool for objectifying and accelerating postmitotic neural migration assays. COMPARISON WITH EXISTING METHODS Until now, conditions for in utero electroporation of C57BL/6 mice are sparsely defined. Further, compared with time-consuming cell body counting histogram analysis allows objectified and accelerated postmitotic neural migration assays. CONCLUSION Together, our results provide a manual for the in utero electroporation of specific regions of the central nervous systems C57BL/6 mice and objectified data analysis.
Collapse
|
42
|
Aloia L, Di Stefano B, Sessa A, Morey L, Santanach A, Gutierrez A, Cozzuto L, Benitah SA, Graf T, Broccoli V, Di Croce L. Zrf1 is required to establish and maintain neural progenitor identity. Genes Dev 2014; 28:182-97. [PMID: 24449271 PMCID: PMC3909791 DOI: 10.1101/gad.228510.113] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
The molecular mechanisms underlying specification from embryonic stem cells (ESCs) and maintenance of neural progenitor cells (NPCs) are largely unknown. Recently, we reported that the Zuotin-related factor 1 (Zrf1) is necessary for chromatin displacement of the Polycomb-repressive complex 1 (PRC1). We found that Zrf1 is required for NPC specification from ESCs and that it promotes the expression of NPC markers, including the key regulator Pax6. Moreover, Zrf1 is essential to establish and maintain Wnt ligand expression levels, which are necessary for NPC self-renewal. Reactivation of proper Wnt signaling in Zrf1-depleted NPCs restores Pax6 expression and the self-renewal capacity. ESC-derived NPCs in vitro resemble most of the characteristics of the self-renewing NPCs located in the developing embryonic cortex, which are termed radial glial cells (RGCs). Depletion of Zrf1 in vivo impairs the expression of key self-renewal regulators and Wnt ligand genes in RGCs. Thus, we demonstrate that Zrf1 plays an essential role in NPC generation and maintenance.
Collapse
Affiliation(s)
- Luigi Aloia
- Centre for Genomic Regulation (CRG), Universitat Pompeu Fabra, 08003 Barcelona, Spain
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Choi J, Park S, Sockanathan S. Activated retinoid receptors are required for the migration and fate maintenance of subsets of cortical neurons. Development 2014; 141:1151-60. [PMID: 24504337 DOI: 10.1242/dev.104505] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Layer-specific cortical neurons are essential components of local, intracortical and subcortical circuits and are specified by complex signaling pathways acting on cortical progenitors. However, whether extrinsic signals contribute to postmitotic cortical neuronal development is unclear. Here we show in mice that retinoic acid (RA) receptors are activated in newly born migrating cortical neurons indicative of endogenous RA in the cortex. Disruption of RA signaling in postmitotic neurons by dominant-negative retinoid receptor RAR403 expression specifically delays late-born cortical neuron migration in vivo. Moreover, prospective layer V-III neurons that express RAR403 fail to maintain their fates and instead acquire characteristics of layer II neurons. This latter phenotype is rescued by active forms of β-catenin at central and caudal but not rostral cortical regions. Taken together, these observations suggest that RA signaling pathways operate postmitotically to regulate the onset of radial migration and to consolidate regional differences in cortical neuronal identity.
Collapse
Affiliation(s)
- Jeonghoon Choi
- PCTB1004, the Solomon Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, 725 North Wolfe Street, Baltimore, MD 21205, USA
| | | | | |
Collapse
|
44
|
Inoue M, Kuroda T, Honda A, Komabayashi-Suzuki M, Komai T, Shinkai Y, Mizutani KI. Prdm8 regulates the morphological transition at multipolar phase during neocortical development. PLoS One 2014; 9:e86356. [PMID: 24489718 PMCID: PMC3906029 DOI: 10.1371/journal.pone.0086356] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2013] [Accepted: 12/06/2013] [Indexed: 12/31/2022] Open
Abstract
Here, we found that the PR domain protein Prdm8 serves as a key regulator of the length of the multipolar phase by controlling the timing of morphological transition. We used a mouse line with expression of Prdm8-mVenus reporter and found that Prdm8 is predominantly expressed in the middle and upper intermediate zone during both the late and terminal multipolar phases. Prdm8 expression was almost coincident with Unc5D expression, a marker for the late multipolar phase, although the expression of Unc5D was found to be gradually down-regulated to the point at which mVenus expression was gradually up-regulated. This expression pattern suggests the possible involvement of Prdm8 in the control of the late and terminal multipolar phases, which controls the timing for morphological transition. To test this hypothesis, we performed gain- and loss-of-function analysis of neocortical development by using in utero electroporation. We found that the knockdown of Prdm8 results in premature change from multipolar to bipolar morphology, whereas the overexpression of Prdm8 maintained the multipolar morphology. Additionally, the postnatal analysis showed that the Prdm8 knockdown stimulated the number of early born neurons, and differentiated neurons located more deeply in the neocortex, however, majority of those cells could not acquire molecular features consistent with laminar location. Furthermore, we found the candidate genes that were predominantly utilized in both the late and terminal multipolar phases, and these candidate genes included those encoding for guidance molecules. In addition, we also found that the expression level of these guidance molecules was inhibited by the introduction of the Prdm8 expression vector. These results indicate that the Prdm8-mediated regulation of morphological changes that normally occur during the late and terminal multipolar phases plays an important role in neocortical development.
Collapse
Affiliation(s)
- Mayuko Inoue
- Laboratory of Neural Differentiation, Graduate School of Brain Science, Doshisha University, Kyoto, Japan
- Department of Molecular and Cellular Biology, Graduate School of Biostudies, Kyoto University, Kyoto, Japan
| | - Takao Kuroda
- Laboratory of Neural Differentiation, Graduate School of Brain Science, Doshisha University, Kyoto, Japan
| | - Aya Honda
- Laboratory of Neural Differentiation, Graduate School of Brain Science, Doshisha University, Kyoto, Japan
| | - Mariko Komabayashi-Suzuki
- Laboratory of Neural Differentiation, Graduate School of Brain Science, Doshisha University, Kyoto, Japan
| | - Tae Komai
- Department of Molecular and Cellular Biology, Graduate School of Biostudies, Kyoto University, Kyoto, Japan
| | | | - Ken-ichi Mizutani
- Laboratory of Neural Differentiation, Graduate School of Brain Science, Doshisha University, Kyoto, Japan
- Japan Science and Technology Agency, PRESTO, Tokyo, Japan
| |
Collapse
|
45
|
Ratié L, Ware M, Barloy-Hubler F, Romé H, Gicquel I, Dubourg C, David V, Dupé V. Novel genes upregulated when NOTCH signalling is disrupted during hypothalamic development. Neural Dev 2013; 8:25. [PMID: 24360028 PMCID: PMC3880542 DOI: 10.1186/1749-8104-8-25] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2013] [Accepted: 12/10/2013] [Indexed: 12/11/2022] Open
Abstract
Background The generation of diverse neuronal types and subtypes from multipotent progenitors during development is crucial for assembling functional neural circuits in the adult central nervous system. It is well known that the Notch signalling pathway through the inhibition of proneural genes is a key regulator of neurogenesis in the vertebrate central nervous system. However, the role of Notch during hypothalamus formation along with its downstream effectors remains poorly defined. Results Here, we have transiently blocked Notch activity in chick embryos and used global gene expression analysis to provide evidence that Notch signalling modulates the generation of neurons in the early developing hypothalamus by lateral inhibition. Most importantly, we have taken advantage of this model to identify novel targets of Notch signalling, such as Tagln3 and Chga, which were expressed in hypothalamic neuronal nuclei. Conclusions These data give essential advances into the early generation of neurons in the hypothalamus. We demonstrate that inhibition of Notch signalling during early development of the hypothalamus enhances expression of several new markers. These genes must be considered as important new targets of the Notch/proneural network.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Valérie Dupé
- Institut de Génétique et Développement de Rennes, CNRS UMR6290, Université de Rennes 1, IFR140 GFAS, Faculté de Médecine, Rennes, France.
| |
Collapse
|
46
|
Sequerra EB, Costa MR, Menezes JRL, Hedin-Pereira C. Adult neural stem cells: plastic or restricted neuronal fates? Development 2013; 140:3303-9. [PMID: 23900539 DOI: 10.1242/dev.093096] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
During embryonic development, the telencephalon is specified along its axis through morphogenetic gradients, leading to the positional-dependent generation of multiple neuronal types. After embryogenesis, however, the fate of neuronal progenitors becomes more restricted, and they generate only a subset of neurons. Here, we review studies of postnatal and adult neurogenesis, challenging the notion that fixed genetic programs restrict neuronal fate. We hypothesize that the adult brain maintains plastic neural stem cells that are capable of responding to changes in environmental cues and generating diverse neuronal types. Thus, the limited diversity of neurons generated under normal conditions must be actively maintained by the adult milieu.
Collapse
Affiliation(s)
- Eduardo B Sequerra
- Department of Physiology and Membrane Biology, University of California Davis, Shriners Hospital for Children Northern California, Sacramento, CA 95817, USA.
| | | | | | | |
Collapse
|
47
|
Boissart C, Poulet A, Georges P, Darville H, Julita E, Delorme R, Bourgeron T, Peschanski M, Benchoua A. Differentiation from human pluripotent stem cells of cortical neurons of the superficial layers amenable to psychiatric disease modeling and high-throughput drug screening. Transl Psychiatry 2013; 3:e294. [PMID: 23962924 PMCID: PMC3756296 DOI: 10.1038/tp.2013.71] [Citation(s) in RCA: 85] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2013] [Revised: 07/19/2013] [Accepted: 07/19/2013] [Indexed: 12/13/2022] Open
Abstract
Cortical neurons of the superficial layers (II-IV) represent a pivotal neuronal population involved in the higher cognitive functions of the human and are particularly affected by psychiatric diseases with developmental manifestations such as schizophrenia and autism. Differentiation protocols of human pluripotent stem cells (PSC) into cortical neurons have been achieved, opening the way to in vitro modeling of neuropsychiatric diseases. However, these protocols commonly result in the asynchronous production of neurons typical for the different layers of the cortex within an extended period of culture, thus precluding the analysis of specific subtypes of neurons in a standardized manner. Addressing this issue, we have successfully captured a stable population of self-renewing late cortical progenitors (LCPs) that synchronously and massively differentiate into glutamatergic cortical neurons of the upper layers. The short time course of differentiation into neurons of these progenitors has made them amenable to high-throughput assays. This has allowed us to analyze the capability of LCPs at differentiating into post mitotic neurons as well as extending and branching neurites in response to a collection of selected bioactive molecules. LCPs and cortical neurons of the upper layers were successfully produced from patient-derived-induced PSC, indicating that this system enables functional studies of individual-specific cortical neurons ex vivo for disease modeling and therapeutic purposes.
Collapse
Affiliation(s)
- C Boissart
- Neuroplasticity and Therapeutics, CECS, I-STEM, AFM, Evry Cedex, France
| | - A Poulet
- Neuroplasticity and Therapeutics, CECS, I-STEM, AFM, Evry Cedex, France
| | - P Georges
- Neuroplasticity and Therapeutics, CECS, I-STEM, AFM, Evry Cedex, France
| | - H Darville
- Neuroplasticity and Therapeutics, CECS, I-STEM, AFM, Evry Cedex, France
| | - E Julita
- Neuroplasticity and Therapeutics, CECS, I-STEM, AFM, Evry Cedex, France
| | - R Delorme
- Human Genetics and Cognitive Functions, Institut Pasteur, Paris, France,Assistance Publique-Hôpitaux de Paris, Robert Debré Hospital, Department of Child and Adolescent Psychiatry, Paris, France
| | - T Bourgeron
- Human Genetics and Cognitive Functions, Institut Pasteur, Paris, France,CNRS URA 2182 ‘Genes, synapses and cognition', Institut Pasteur, Paris, France,University Denis Diderot Paris 7, Paris, France
| | - M Peschanski
- INSERM/UEVE UMR 861 I-STEM AFM, Evry Cedex, France
| | - A Benchoua
- Neuroplasticity and Therapeutics, CECS, I-STEM, AFM, Evry Cedex, France,Neuroplasticity and Therapeutics, CECS, I-STEM, AFM, 5 rue Henri Desbrueres, Genopole campus 1, Evry Cedex 91030, France. E-mail:
| |
Collapse
|
48
|
Shih HP, Wang A, Sander M. Pancreas organogenesis: from lineage determination to morphogenesis. Annu Rev Cell Dev Biol 2013; 29:81-105. [PMID: 23909279 DOI: 10.1146/annurev-cellbio-101512-122405] [Citation(s) in RCA: 222] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
The pancreas is an essential organ for proper nutrient metabolism and has both endocrine and exocrine function. In the past two decades, knowledge of how the pancreas develops during embryogenesis has significantly increased, largely from developmental studies in model organisms. Specifically, the molecular basis of pancreatic lineage decisions and cell differentiation is well studied. Still not well understood are the mechanisms governing three-dimensional morphogenesis of the organ. Strategies to derive transplantable β-cells in vitro for diabetes treatment have benefited from the accumulated knowledge of pancreas development. In this review, we provide an overview of the current understanding of pancreatic lineage determination and organogenesis, and we examine future implications of these findings for treatment of diabetes mellitus through cell replacement.
Collapse
Affiliation(s)
- Hung Ping Shih
- Departments of Pediatrics and Cellular & Molecular Medicine, Pediatric Diabetes Research Center, University of California, San Diego, La Jolla, California 92093-0695;
| | | | | |
Collapse
|
49
|
Aujla PK, Naratadam GT, Xu L, Raetzman LT. Notch/Rbpjκ signaling regulates progenitor maintenance and differentiation of hypothalamic arcuate neurons. Development 2013; 140:3511-21. [PMID: 23884446 DOI: 10.1242/dev.098681] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
The hypothalamic arcuate nucleus (Arc), containing pro-opoiomelanocortin (POMC), neuropeptide Y (NPY) and growth hormone releasing hormone (GHRH) neurons, regulates feeding, energy balance and body size. Dysregulation of this homeostatic mediator underlies diseases ranging from growth failure to obesity. Despite considerable investigation regarding the function of Arc neurons, mechanisms governing their development remain unclear. Notch signaling factors such as Hes1 and Mash1 are present in hypothalamic progenitors that give rise to Arc neurons. However, how Notch signaling controls these progenitor populations is unknown. To elucidate the role of Notch signaling in Arc development, we analyzed conditional loss-of-function mice lacking a necessary Notch co-factor, Rbpjκ, in Nkx2.1-cre-expressing cells (Rbpjκ cKO), as well as mice with expression of the constitutively active Notch1 intracellular domain (NICD) in Nkx2.1-cre-expressing cells (NICD Tg). We found that loss of Rbpjκ results in absence of Hes1 but not of Hes5 within the primordial Arc at E13.5. Additionally, Mash1 expression is increased, coincident with increased proliferation and accumulation of Arc neurons at E13.5. At E18.5, Rbpjκ cKO mice have few progenitors and show increased numbers of differentiated Pomc, NPY and Ghrh neurons. By contrast, NICD Tg mice have increased hypothalamic progenitors, show an absence of differentiated Arc neurons and aberrant glial differentiation at E18.5. Subsequently, both Rbpjκ cKO and NICD Tg mice have changes in growth and body size during postnatal development. Taken together, our results demonstrate that Notch/Rbpjκ signaling regulates the generation and differentiation of Arc neurons, which contribute to homeostatic regulation of body size.
Collapse
Affiliation(s)
- Paven K Aujla
- University of Illinois at Urbana-Champaign, 407 South Goodwin Avenue, Urbana, IL 61801, USA
| | | | | | | |
Collapse
|
50
|
Shimada JI, Taniguchi J, Mori M, Sato Y, Takuwa H, Ito H, Kuwabara S. Retinol palmitate prevents ischemia-induced cell changes in hippocampal neurons through the Notch1 signaling pathway in mice. Exp Neurol 2013; 247:182-7. [PMID: 23651513 DOI: 10.1016/j.expneurol.2013.04.014] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2012] [Revised: 04/23/2013] [Accepted: 04/29/2013] [Indexed: 02/05/2023]
Abstract
Retinol palmitate, an analog of vitamin A, plays multiple roles in the nervous system, including neural differentiation, axon outgrowth, and neural patterning, and is also an antioxidative agent and thereby potential neuroprotectant for brain ischemia. The present study aimed at investigating the protective effects of retinol palmitate against ischemia-induced brain injury in a bilateral common carotid artery occlusion (BCCAO) model in mice. Ischemia induced by 20-min BCCAO resulted in significant neuronal morphological changes and reactive astrocyte proliferation in the hippocampus, particularly in the CA1 region, and these changes were accompanied by increased Notch1 expression. Intraperitoneal retinol palmitate administration before ischemia reduced ischemic neurons with Notch1 expression; the differences were statistically significant in both the 1.2mg/kg group and 12 mg/kg group. These results show that retinol palmitate prevents brain ischemia-induced neuronal injury with Notch1 expression and that Notch1 signaling could be involved in the neuroprotective mechanism. Retinol palmitate could be a treatment option for human brain infarction.
Collapse
Affiliation(s)
- Jun-Ichiro Shimada
- Department of Neurology, Graduate School of Medicine, Chiba University, Chuo-ku, Chiba 260-8670, Japan
| | | | | | | | | | | | | |
Collapse
|