51
|
Genetic and Molecular Approaches to Study Neuronal Migration in the Developing Cerebral Cortex. Brain Sci 2017; 7:brainsci7050053. [PMID: 28475113 PMCID: PMC5447935 DOI: 10.3390/brainsci7050053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2017] [Revised: 04/21/2017] [Accepted: 05/02/2017] [Indexed: 11/17/2022] Open
Abstract
The migration of neuronal cells in the developing cerebral cortex is essential for proper development of the brain and brain networks. Disturbances in this process, due to genetic abnormalities or exogenous factors, leads to aberrant brain formation, brain network formation, and brain function. In the last decade, there has been extensive research in the field of neuronal migration. In this review, we describe different methods and approaches to assess and study neuronal migration in the developing cerebral cortex. First, we discuss several genetic methods, techniques and genetic models that have been used to study neuronal migration in the developing cortex. Second, we describe several molecular approaches to study aberrant neuronal migration in the cortex which can be used to elucidate the underlying mechanisms of neuronal migration. Finally, we describe model systems to investigate and assess the potential toxicity effect of prenatal exposure to environmental chemicals on proper brain formation and neuronal migration.
Collapse
|
52
|
Kim J, Yang K, Lee JS, Hwang YH, Park HJ, Park KI, Lee DY, Cho SW. Enhanced Self-Renewal and Accelerated Differentiation of Human Fetal Neural Stem Cells Using Graphene Oxide Nanoparticles. Macromol Biosci 2017; 17. [PMID: 28394476 DOI: 10.1002/mabi.201600540] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2016] [Revised: 02/26/2017] [Indexed: 01/09/2023]
Abstract
Graphene oxide (GO) has received increasing attention in bioengineering fields due to its unique biophysical and electrical properties, along with excellent biocompatibility. The application of GO nanoparticles (GO-NPs) to engineer self-renewal and differentiation of human fetal neural stem cells (hfNSCs) is reported. GO-NPs added to hfNSC culture during neurosphere formation substantially promote cell-to-cell and cell-to-matrix interactions in neurospheres. Accordingly, GO-NP-treated hfNSCs show enhanced self-renewal ability and accelerated differentiation compared to untreated cells, indicating the utility of GO in developing stem cell therapies for neurogenesis.
Collapse
Affiliation(s)
- Jin Kim
- Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
| | - Kisuk Yang
- Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
| | - Jong Seung Lee
- Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
| | - Yong Hwa Hwang
- Department of Bioengineering, College of Engineering, and BK21 PLUS Future Biopharmaceutical Human Resources Training and Research Team, and Institute of Nano Science and Technology (INST), Hanyang University, Seoul, 04763, Republic of Korea
| | - Hyun-Ji Park
- Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
| | - Kook In Park
- Severance Children's Hospital, Department of Pediatrics and BK21 Plus Project for Medical Sciences, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
| | - Dong Yun Lee
- Department of Bioengineering, College of Engineering, and BK21 PLUS Future Biopharmaceutical Human Resources Training and Research Team, and Institute of Nano Science and Technology (INST), Hanyang University, Seoul, 04763, Republic of Korea
| | - Seung-Woo Cho
- Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
| |
Collapse
|
53
|
Maki T. Novel roles of oligodendrocyte precursor cells in the developing and damaged brain. ACTA ACUST UNITED AC 2017. [DOI: 10.1111/cen3.12358] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
- Takakuni Maki
- Department of Neurology; Graduate School of Medicine; Kyoto University; Kyoto Japan
| |
Collapse
|
54
|
Huntingtin-Mediated Multipolar-Bipolar Transition of Newborn Cortical Neurons Is Critical for Their Postnatal Neuronal Morphology. Neuron 2017; 93:99-114. [DOI: 10.1016/j.neuron.2016.11.035] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2016] [Revised: 09/25/2016] [Accepted: 11/16/2016] [Indexed: 01/05/2023]
|
55
|
Chou FS, Wang PS. The Arp2/3 complex is essential at multiple stages of neural development. NEUROGENESIS 2016; 3:e1261653. [PMID: 28405589 DOI: 10.1080/23262133.2016.1261653] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/23/2016] [Revised: 11/02/2016] [Accepted: 11/11/2016] [Indexed: 12/20/2022]
Abstract
During development of the nervous system, radial glial cells perform self-renewing asymmetric divisions and give rise to intermediate progenitor cells (IPC) and neurons. The neuronally committed IPC subsequently undergo multiple rounds of transient amplification and migrate outwards to form cortical layers as they continue to differentiate into mature neurons. Maturing neurons extend protrusions on their cell surface to form neurites, a process called neuritogenesis. Neurite formation results in the establishment of dendrites and axons for synapse formation, which is essential for sensory and motor functions and even higher-level functioning including memory formation and cognitive function, as well as shaping of behavior and emotion. Morphological adaptation during various stages of neural development requires active participation of actin cytoskeleton remodeling. In this review, we aim to discuss current understanding of the Arp2/3 complex branching nucleator in various neural cell types during development and maturation.
Collapse
Affiliation(s)
- Fu-Sheng Chou
- Department of Pediatrics, University of Kansas Medical Center, Kansas City, KS, USA; Department of Pediatrics, University of Missouri-Kansas City, Kansas City, MO, USA; Section of Neonatology, Children's Mercy-Kansas City, Kansas City, MO, USA
| | - Pei-Shan Wang
- Department of Pediatrics, University of Kansas Medical Center, Kansas City, KS, USA; Department of Pediatrics, University of Missouri-Kansas City, Kansas City, MO, USA
| |
Collapse
|
56
|
Yang C, Li X, Wang C, Fu S, Li H, Guo Z, Zhao S, Lin J. N-cadherin regulates beta-catenin signal and its misexpression perturbs commissural axon projection in the developing chicken spinal cord. J Mol Histol 2016; 47:541-554. [PMID: 27650519 DOI: 10.1007/s10735-016-9698-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2016] [Accepted: 09/14/2016] [Indexed: 12/31/2022]
Abstract
N-cadherin is a calcium-sensitive cell adhesion molecule that plays an important role in the formation of the neural circuit and the development of the nervous system. In the present study, we investigated the function of N-cadherin in cell-cell connection in vitro with HEK293T cells, and in commissural axon projections in the developing chicken spinal cord using in ovo electroporation. Cell-cell connections increased with N-cadherin overexpression in HEK293T cells, while cell contacts disappeared after co-transfection with an N-cadherin-shRNA plasmid. The knockdown of N-cadherin caused the accumulation of β-catenin in the nucleus, supporting the notion that N-cadherin regulates β-catenin signaling in vitro. Furthermore, N-cadherin misexpression perturbed commissural axon projections in the spinal cord. The overexpression of N-cadherin reduced the number of axons that projected alongside the contralateral margin of the floor plate, and formed intermediate longitudinal commissural axons. In contrast, the knockdown of N-cadherin perturbed commissural axon projections significantly, affecting the projections alongside the contralateral margin of the floor plate, but did not affect intermediate longitudinal commissural axons. Taken together, these findings suggest that N-cadherin regulates commissural axon projections in the developing chicken spinal cord.
Collapse
Affiliation(s)
- Ciqing Yang
- College of Life Science and Technology, Xinxiang Medical University, Xinxiang, 453003, China.,Henan Key Laboratory of Medical Tissue Regeneration, Xinxiang, 453003, China
| | - Xiaoying Li
- College of Life Science and Technology, Xinxiang Medical University, Xinxiang, 453003, China
| | - Congrui Wang
- Henan Key Laboratory of Medical Tissue Regeneration, Xinxiang, 453003, China
| | - Sulei Fu
- College of Life Science and Technology, Xinxiang Medical University, Xinxiang, 453003, China
| | - Han Li
- College of Life Science and Technology, Xinxiang Medical University, Xinxiang, 453003, China.,Advanced Medical and Dental Institute, Universiti Sains Malaysia, 13200, Bertam, Penang, Malaysia
| | - Zhikun Guo
- Henan Key Laboratory of Medical Tissue Regeneration, Xinxiang, 453003, China
| | - Shanting Zhao
- College of Veterinary Medicine, Northwest A&F University, Yangling, 712100, China
| | - Juntang Lin
- College of Life Science and Technology, Xinxiang Medical University, Xinxiang, 453003, China. .,Henan Key Laboratory of Medical Tissue Regeneration, Xinxiang, 453003, China. .,Institute of Anatomy I, Jena University Hospital, 07743, Jena, Germany.
| |
Collapse
|
57
|
Matsuda M, Rand K, Palardy G, Shimizu N, Ikeda H, Dalle Nogare D, Itoh M, Chitnis AB. Epb41l5 competes with Delta as a substrate for Mib1 to coordinate specification and differentiation of neurons. Development 2016; 143:3085-96. [PMID: 27510968 DOI: 10.1242/dev.138743] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2016] [Accepted: 07/26/2016] [Indexed: 01/04/2023]
Abstract
We identified Erythrocyte membrane protein band 4.1-like 5 (Epb41l5) as a substrate for the E3 ubiquitin ligase Mind bomb 1 (Mib1), which is essential for activation of Notch signaling. Although loss of Epb41l5 does not significantly alter the pattern of neural progenitor cells (NPCs) specified as neurons at the neural plate stage, it delays their delamination and differentiation after neurulation when NPCs normally acquire organized apical junctional complexes (AJCs) in the zebrafish hindbrain. Delays in differentiation are reduced by knocking down N-cadherin, a manipulation expected to help destabilize adherens junctions (AJs). This suggested that delays in neuronal differentiation in epb41l5-deficient embryos are related to a previously described role for Epb41l5 in facilitating disassembly of cadherin-dependent AJCs. Mib1 ubiquitylates Epb41l5 to promote its degradation. DeltaD can compete with Epb41l5 to reduce Mib1-dependent Epb41l5 degradation. In this context, increasing the number of NPCs specified to become neurons, i.e. cells expressing high levels of DeltaD, stabilizes Epb41l5 in the embryo. Together, these observations suggest that relatively high levels of Delta stabilize Epb41l5 in NPCs specified as neurons. This, we suggest, helps coordinate NPC specification with Epb41l5-dependent delamination and differentiation as neurons.
Collapse
Affiliation(s)
- Miho Matsuda
- Department of Cell Biology and Molecular Medicine, Rutgers, The State University of New Jersey, New Jersey Medical School, Newark, NJ 07103, USA Section on Neural Developmental Dynamics, Division of Developmental Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA
| | - Kinneret Rand
- Section on Neural Developmental Dynamics, Division of Developmental Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA
| | - Greg Palardy
- Section on Neural Developmental Dynamics, Division of Developmental Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA
| | - Nobuyuki Shimizu
- Department of Cell Biology and Molecular Medicine, Rutgers, The State University of New Jersey, New Jersey Medical School, Newark, NJ 07103, USA
| | - Hiromi Ikeda
- Section on Neural Developmental Dynamics, Division of Developmental Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA
| | - Damian Dalle Nogare
- Section on Neural Developmental Dynamics, Division of Developmental Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA
| | - Motoyuki Itoh
- Section on Neural Developmental Dynamics, Division of Developmental Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA Department of Pharmacology, Chiba University, Chiba 260-8675, Japan
| | - Ajay B Chitnis
- Section on Neural Developmental Dynamics, Division of Developmental Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
58
|
Wang PS, Chou FS, Ramachandran S, Xia S, Chen HY, Guo F, Suraneni P, Maher BJ, Li R. Crucial roles of the Arp2/3 complex during mammalian corticogenesis. Development 2016; 143:2741-52. [PMID: 27385014 PMCID: PMC5004905 DOI: 10.1242/dev.130542] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2015] [Accepted: 06/20/2016] [Indexed: 02/06/2023]
Abstract
The polarity and organization of radial glial cells (RGCs), which serve as both stem cells and scaffolds for neuronal migration, are crucial for cortical development. However, the cytoskeletal mechanisms that drive radial glial outgrowth and maintain RGC polarity remain poorly understood. Here, we show that the Arp2/3 complex – the unique actin nucleator that produces branched actin networks – plays essential roles in RGC polarity and morphogenesis. Disruption of the Arp2/3 complex in murine RGCs retards process outgrowth toward the basal surface and impairs apical polarity and adherens junctions. Whereas the former is correlated with an abnormal actin-based leading edge, the latter is consistent with blockage in membrane trafficking. These defects result in altered cell fate, disrupted cortical lamination and abnormal angiogenesis. In addition, we present evidence that the Arp2/3 complex is a cell-autonomous regulator of neuronal migration. Our data suggest that Arp2/3-mediated actin assembly might be particularly important for neuronal cell motility in a soft or poorly adhesive matrix environment. Summary: During mouse cortical development, the Arp2/3 actin branching complex regulates process formation and the maintenance of radial glial cell polarity, as well as affecting neuronal migration.
Collapse
Affiliation(s)
- Pei-Shan Wang
- Stowers Institute for Medical Research, 1000 East 50th Street, Kansas City, MO 64110, USA
| | - Fu-Sheng Chou
- Division of Newborn Medicine, Boston Children's Hospital, Harvard Medical School, 300 Longwood Ave., Boston, MA 02115, USA
| | - Sreekumar Ramachandran
- Department of Cell Biology, Johns Hopkins University School of Medicine, 855 North Wolfe Street, Baltimore, MD 21205, USA
| | - Sheng Xia
- Department of Cell Biology, Johns Hopkins University School of Medicine, 855 North Wolfe Street, Baltimore, MD 21205, USA
| | - Huei-Ying Chen
- Lieber Institute for Brain Development, 855 North Wolfe Street, Baltimore, MD 21205, USA
| | - Fengli Guo
- Stowers Institute for Medical Research, 1000 East 50th Street, Kansas City, MO 64110, USA
| | - Praveen Suraneni
- Division of Hematology/Oncology, Robert Lurie Comprehensive Cancer Center, Department of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Brady J Maher
- Lieber Institute for Brain Development, 855 North Wolfe Street, Baltimore, MD 21205, USA Department of Psychiatry and Behavioral Sciences, Johns Hopkins School of Medicine, 4940 Eastern Ave., Baltimore, MD 21224, USA Department of Neuroscience, Johns Hopkins School of Medicine, 725 N. Wolfe St., Baltimore, MD 21205, USA
| | - Rong Li
- Stowers Institute for Medical Research, 1000 East 50th Street, Kansas City, MO 64110, USA Department of Cell Biology, Johns Hopkins University School of Medicine, 855 North Wolfe Street, Baltimore, MD 21205, USA Department of Chemical and Biomolecular Engineering, Johns Hopkins University, 3400 N Charles St., Baltimore, MD 21218, USA
| |
Collapse
|
59
|
Crucial Role of Rapgef2 and Rapgef6, a Family of Guanine Nucleotide Exchange Factors for Rap1 Small GTPase, in Formation of Apical Surface Adherens Junctions and Neural Progenitor Development in the Mouse Cerebral Cortex. eNeuro 2016; 3:eN-NWR-0142-16. [PMID: 27390776 PMCID: PMC4917737 DOI: 10.1523/eneuro.0142-16.2016] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2016] [Accepted: 06/04/2016] [Indexed: 12/19/2022] Open
Abstract
Cerebral neocortex development in mammals requires highly orchestrated events involving proliferation, differentiation, and migration of neural progenitors and neurons. Rapgef2 and Rapgef6 constitute a unique family of guanine nucleotide exchange factors for Rap1 small GTPase, which is known to play crucial roles in migration of postmitotic neurons. We previously reported that conditional knockout of Rapgef2 in dorsal telencephalon (Rapgef2-cKO) resulted in the formation of an ectopic cortical mass (ECM) resembling that of subcortical band heterotopia. Here we show that double knockout of Rapgef6 in Rapgef2-cKO mice (Rapgef2/6-dKO) results in marked enlargement of the ECM. While Rapgef2-cKO affects late-born neurons only, Rapgef2/6-dKO affects both early-born and late-born neurons. The Rapgef2-cKO cortex at embryonic day (E) 15.5, and the Rapgef2/6-dKO cortex at E13.5 and E15.5 show disruption of the adherens junctions (AJs) on the apical surface, detachment of radial glial cells (RGCs) from the apical surface and disorganization of the radial glial fiber system, which are accompanied by aberrant distribution of RGCs and intermediate progenitors, normally located in the ventricular zone and the subventricular zone, respectively, over the entire cerebral cortex. Moreover, intrauterine transduction of Cre recombinase into the Rapgef2flox/flox brains also results in the apical surface AJ disruption and the RGC detachment from the apical surface, both of which are effectively suppressed by cotransduction of the constitutively active Rap1 mutant Rap1G12V. These results demonstrate a cell-autonomous role of the Rapgef2/6-Rap1 pathway in maintaining the apical surface AJ structures, which is necessary for the proper development of neural progenitor cells.
Collapse
|
60
|
Liang L, Sun H, Zhang W, Zhang M, Yang X, Kuang R, Zheng H. Meta-Analysis of EMT Datasets Reveals Different Types of EMT. PLoS One 2016; 11:e0156839. [PMID: 27258544 PMCID: PMC4892621 DOI: 10.1371/journal.pone.0156839] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2016] [Accepted: 05/22/2016] [Indexed: 11/25/2022] Open
Abstract
As a critical process during embryonic development, cancer progression and cell fate conversions, epithelial-mesenchymal transition (EMT) has been extensively studied over the last several decades. To further understand the nature of EMT, we performed meta-analysis of multiple microarray datasets to identify the related generic signature. In this study, 24 human and 17 mouse microarray datasets were integrated to identify conserved gene expression changes in different types of EMT. Our integrative analysis revealed that there is low agreement among the list of the identified signature genes and three other lists in previous studies. Since removing the datasets with weakly-induced EMT from the analysis did not significantly improve the overlapping in the signature-gene lists, we hypothesized the existence of different types of EMT. This hypothesis was further supported by the grouping of 74 human EMT-induction samples into five distinct clusters, and the identification of distinct pathways in these different clusters of EMT samples. The five clusters of EMT-induction samples also improves the understanding of the characteristics of different EMT types. Therefore, we concluded the existence of different types of EMT was the possible reason for its complex role in multiple biological processes.
Collapse
Affiliation(s)
- Lining Liang
- CAS Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, Guangdong, China
| | - Hao Sun
- CAS Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, Guangdong, China
| | - Wei Zhang
- Department of Computer Science and Engineering, University of Minnesota Twin Cities, Minneapolis, Minnesota, United States of America
| | - Mengdan Zhang
- CAS Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, Guangdong, China
| | - Xiao Yang
- CAS Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, Guangdong, China
| | - Rui Kuang
- Department of Computer Science and Engineering, University of Minnesota Twin Cities, Minneapolis, Minnesota, United States of America
| | - Hui Zheng
- CAS Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, Guangdong, China
| |
Collapse
|
61
|
Wen MH, Wang JY, Chiu YT, Wang MP, Lee SP, Tai CY. N-Cadherin Regulates Cell Migration Through a Rab5-Dependent Temporal Control of Macropinocytosis. Traffic 2016; 17:769-85. [DOI: 10.1111/tra.12402] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2015] [Revised: 04/06/2016] [Accepted: 04/06/2016] [Indexed: 11/27/2022]
Affiliation(s)
- Meng-Hsuan Wen
- Graduate Institute of Life Sciences; National Defense Medical Center; Taipei 114 Taiwan
- Institute of Molecular Biology; Academia Sinica; Taipei 115 Taiwan
| | - Jen-Yeu Wang
- Institute of Molecular Biology; Academia Sinica; Taipei 115 Taiwan
| | - Yu-Ting Chiu
- Institute of Molecular Biology; Academia Sinica; Taipei 115 Taiwan
- Institute of Genomics Sciences; National Yang-Ming University; Taipei 112 Taiwan
| | - Mei-Pin Wang
- Institute of Molecular Biology; Academia Sinica; Taipei 115 Taiwan
- Institute of Pharmaceutics; Development Center for Biotechnology; New Taipei City 221 Taiwan
| | - Sue-Ping Lee
- Institute of Molecular Biology; Academia Sinica; Taipei 115 Taiwan
| | - Chin-Yin Tai
- Graduate Institute of Life Sciences; National Defense Medical Center; Taipei 114 Taiwan
- Institute of Molecular Biology; Academia Sinica; Taipei 115 Taiwan
- Institute of Genomics Sciences; National Yang-Ming University; Taipei 112 Taiwan
- Institute of Pharmaceutics; Development Center for Biotechnology; New Taipei City 221 Taiwan
| |
Collapse
|
62
|
Egusa SF, Inoue YU, Asami J, Terakawa YW, Hoshino M, Inoue T. Classic cadherin expressions balance postnatal neuronal positioning and dendrite dynamics to elaborate the specific cytoarchitecture of the mouse cortical area. Neurosci Res 2016; 105:49-64. [DOI: 10.1016/j.neures.2015.09.006] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2015] [Revised: 09/20/2015] [Accepted: 09/24/2015] [Indexed: 11/25/2022]
|
63
|
Kornilov SA, Rakhlin N, Koposov R, Lee M, Yrigollen C, Caglayan AO, Magnuson JS, Mane S, Chang JT, Grigorenko EL. Genome-Wide Association and Exome Sequencing Study of Language Disorder in an Isolated Population. Pediatrics 2016; 137:peds.2015-2469. [PMID: 27016271 PMCID: PMC4811310 DOI: 10.1542/peds.2015-2469] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/26/2016] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND AND OBJECTIVE Developmental language disorder (DLD) is a highly prevalent neurodevelopmental disorder associated with negative outcomes in different domains; the etiology of DLD is unknown. To investigate the genetic underpinnings of DLD, we performed genome-wide association and whole exome sequencing studies in a geographically isolated population with a substantially elevated prevalence of the disorder (ie, the AZ sample). METHODS DNA samples were collected from 359 individuals for the genome-wide association study and from 12 severely affected individuals for whole exome sequencing. Multifaceted phenotypes, representing major domains of expressive language functioning, were derived from collected speech samples. RESULTS Gene-based analyses revealed a significant association between SETBP1 and complexity of linguistic output (P = 5.47 × 10(-7)). The analysis of exome variants revealed coding sequence variants in 14 genes, most of which play a role in neural development. Targeted enrichment analysis implicated myocyte enhancer factor-2 (MEF2)-regulated genes in DLD in the AZ population. The main findings were successfully replicated in an independent cohort of children at risk for related disorders (n = 372). CONCLUSIONS MEF2-regulated pathways were identified as potential candidate pathways in the etiology of DLD. Several genes (including the candidate SETBP1 and other MEF2-related genes) seem to jointly influence certain, but not all, facets of the DLD phenotype. Even when genetic and environmental diversity is reduced, DLD is best conceptualized as etiologically complex. Future research should establish whether the signals detected in the AZ population can be replicated in other samples and languages and provide further characterization of the identified pathway.
Collapse
Affiliation(s)
- Sergey A. Kornilov
- Child Study Center, School of Medicine, Yale University, New Haven, Connecticut;,Department of Psychology, University of Connecticut, Storrs, Connecticut;,Haskins Laboratories, New Haven, Connecticut;,Department of Psychology, Moscow State University, Moscow, Russia;,Department of Psychology, Saint Petersburg State University, Saint Petersburg, Russia
| | - Natalia Rakhlin
- Child Study Center, School of Medicine, Yale University, New Haven, Connecticut;,Department of Communication Sciences and Disorders, Wayne State University, Detroit, Michigan
| | - Roman Koposov
- Regional Centre for Child and Youth Mental Health and Child Welfare, UiT The Arctic University of Norway, Tromsø, Norway
| | - Maria Lee
- Child Study Center, School of Medicine, Yale University, New Haven, Connecticut
| | - Carolyn Yrigollen
- The Children's Hospital of Philadelphia, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Ahmet Okay Caglayan
- Child Study Center, School of Medicine, Yale University, New Haven, Connecticut;,Department of Medical Genetics, Istanbul Bilim University, Istanbul, Turkey; and
| | - James S. Magnuson
- Department of Psychology, University of Connecticut, Storrs, Connecticut;,Haskins Laboratories, New Haven, Connecticut
| | - Shrikant Mane
- Child Study Center, School of Medicine, Yale University, New Haven, Connecticut
| | - Joseph T. Chang
- Child Study Center, School of Medicine, Yale University, New Haven, Connecticut
| | - Elena L. Grigorenko
- Child Study Center, School of Medicine, Yale University, New Haven, Connecticut;,Haskins Laboratories, New Haven, Connecticut;,Department of Psychology, Saint Petersburg State University, Saint Petersburg, Russia;,Moscow State University for Psychology and Education, Moscow, Russia
| |
Collapse
|
64
|
Bertrand V. β-catenin-driven binary cell fate decisions in animal development. WILEY INTERDISCIPLINARY REVIEWS-DEVELOPMENTAL BIOLOGY 2016; 5:377-88. [PMID: 26952169 PMCID: PMC5069452 DOI: 10.1002/wdev.228] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/14/2015] [Revised: 12/06/2015] [Accepted: 12/29/2015] [Indexed: 02/05/2023]
Abstract
The Wnt/β‐catenin pathway plays key roles during animal development. In several species, β‐catenin is used in a reiterative manner to regulate cell fate diversification between daughter cells following division. This binary cell fate specification mechanism has been observed in animals that belong to very diverse phyla: the nematode Caenorhabditis elegans, the annelid Platynereis, and the ascidian Ciona. It may also play a role in the regulation of several stem cell lineages in vertebrates. While the molecular mechanism behind this binary cell fate switch is not fully understood, it appears that both secreted Wnt ligands and asymmetric cortical factors contribute to the generation of the difference in nuclear β‐catenin levels between daughter cells. β‐Catenin then cooperates with lineage specific transcription factors to induce the expression of novel sets of transcription factors at each round of divisions, thereby diversifying cell fate. WIREs Dev Biol 2016, 5:377–388. doi: 10.1002/wdev.228 For further resources related to this article, please visit the WIREs website.
Collapse
Affiliation(s)
- Vincent Bertrand
- Aix-Marseille Université, CNRS, Institut de Biologie du Développement de Marseille, Marseille, France
| |
Collapse
|
65
|
Deglincerti A, Etoc F, Ozair MZ, Brivanlou AH. Self-Organization of Spatial Patterning in Human Embryonic Stem Cells. Curr Top Dev Biol 2016; 116:99-113. [PMID: 26970615 DOI: 10.1016/bs.ctdb.2015.11.010] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
The developing embryo is a remarkable example of self-organization, where functional units are created in a complex spatiotemporal choreography. Recently, human embryonic stem cells (ESCs) have been used to recapitulate in vitro the self-organization programs that are executed in the embryo in vivo. This represents an unique opportunity to address self-organization in humans that is otherwise not addressable with current technologies. In this chapter, we review the recent literature on self-organization of human ESCs, with a particular focus on two examples: formation of embryonic germ layers and neural rosettes. Intriguingly, both activation and elimination of TGFβ signaling can initiate self-organization, albeit with different molecular underpinnings. We discuss the mechanisms underlying the formation of these structures in vitro and explore future challenges in the field.
Collapse
Affiliation(s)
- Alessia Deglincerti
- Laboratory of Stem Cell Biology and Molecular Embryology, The Rockefeller University, New York, USA
| | - Fred Etoc
- Laboratory of Stem Cell Biology and Molecular Embryology, The Rockefeller University, New York, USA
| | - M Zeeshan Ozair
- Laboratory of Stem Cell Biology and Molecular Embryology, The Rockefeller University, New York, USA
| | - Ali H Brivanlou
- Laboratory of Stem Cell Biology and Molecular Embryology, The Rockefeller University, New York, USA.
| |
Collapse
|
66
|
|
67
|
Abstract
The disproportional enlargement of the neocortex through evolution has been instrumental in the success of vertebrates, in particular mammals. The neocortex is a multilayered sheet of neurons generated from a simple proliferative neuroepithelium through a myriad of mechanisms with substantial evolutionary conservation. This developing neuroepithelium is populated by progenitors that can generate additional progenitors as well as post-mitotic neurons. Subtle alterations in the production of progenitors vs. differentiated cells during development can result in dramatic differences in neocortical size. This review article will examine how cadherin adhesion proteins, in particular α-catenin and N-cadherin, function in regulating the neural progenitor microenvironment, cell proliferation, and differentiation in cortical development.
Collapse
Key Words
- APC, Adenomatous polyposis coli.
- CBD, catenin binding domain
- CK1, Casein kinase 1
- GSK3β, glycogen synthase kinase 3β
- Hh, Hedgehog
- JMD, juxtamembrane domain
- N-cadherin
- PCP, planar cell polarity
- PI3K, phosphatidylinositol 3-kinase
- PTEN, phosphatase and tensin homolog
- SHH, sonic hedgehog
- SNP, short neural precursor
- VZ, ventricular zone
- adherens junction
- differentiation
- proliferation
- wnt
- α-catenin
- β-catenin
Collapse
Affiliation(s)
- Adam M Stocker
- a Molecular Neurobiology Laboratory ; The Salk Institute ; La Jolla , CA USA
| | | |
Collapse
|
68
|
Chen D, Wu Z, Luo LJ, Huang X, Qian WQ, Wang H, Li SH, Liu J. E-cadherin maintains the activity of neural stem cells and inhibits the migration. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2015; 8:14247-14251. [PMID: 26823740 PMCID: PMC4713526] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 09/28/2015] [Accepted: 10/28/2015] [Indexed: 06/05/2023]
Abstract
Cadherins are important adhesion molecules that mediate adhesions and communications between cells. These molecules participate in the formation and maintenance of multicellular organisms including the stem cells. E-cadherin is one of the classic cadherins which is reported to be essential for the survival and self-renewal of embryonic stem cells. Moreover, it could induce cell proliferation inhibitory signaling to regulate cell proliferation. In our study, we over-expressed and silenced E-cadherin in NSCs by lentiviral ways. Transgenic cells were confirmed by both quantitative RT-PCR and western blot. Results of MTT assay showed that over-expression of E-cadherin could enhance the cell activity. Furthermore, we performed Transwell chamber assay to analyze its role in regulation of cell migration. The results showed that the migration percent of over-expression cells was lower than control. Our results indicated that E-caherin would maintain the stemness of NSCs and reduce cells migration.
Collapse
Affiliation(s)
- Dong Chen
- Department of Orthopedics, Shanghai Tenth People's Hospital, School of Medicine, Tongji University Shanghai, China
| | - Zhong Wu
- Department of Orthopedics, Shanghai Tenth People's Hospital, School of Medicine, Tongji University Shanghai, China
| | - Lin-Jie Luo
- Department of Orthopedics, Shanghai Tenth People's Hospital, School of Medicine, Tongji University Shanghai, China
| | - Xiang Huang
- Department of Orthopedics, Shanghai Tenth People's Hospital, School of Medicine, Tongji University Shanghai, China
| | - Wei-Qiang Qian
- Department of Orthopedics, Shanghai Tenth People's Hospital, School of Medicine, Tongji University Shanghai, China
| | - Hua Wang
- Department of Orthopedics, Shanghai Tenth People's Hospital, School of Medicine, Tongji University Shanghai, China
| | - Shao-Hua Li
- Department of Orthopedics, Shanghai Tenth People's Hospital, School of Medicine, Tongji University Shanghai, China
| | - Jie Liu
- Department of Orthopedics, Shanghai Tenth People's Hospital, School of Medicine, Tongji University Shanghai, China
| |
Collapse
|
69
|
Bengoa-Vergniory N, Kypta RM. Canonical and noncanonical Wnt signaling in neural stem/progenitor cells. Cell Mol Life Sci 2015; 72:4157-72. [PMID: 26306936 PMCID: PMC11113751 DOI: 10.1007/s00018-015-2028-6] [Citation(s) in RCA: 119] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2015] [Revised: 07/17/2015] [Accepted: 08/18/2015] [Indexed: 02/07/2023]
Abstract
The first mammalian Wnt to be discovered, Wnt-1, was found to be essential for the development of a large part of the mouse brain over 25 years ago. We have since learned that Wnt family secreted glycolipoproteins, of which there are nineteen, which activate a diverse network of signals that are particularly important during embryonic development and tissue regeneration. Wnt signals in the developing and adult brain can drive neural stem cell self-renewal, expansion, asymmetric cell division, maturation and differentiation. The molecular events taking place after a Wnt binds to its cell-surface receptors are complex and, at times, controversial. A deeper understanding of these events is anticipated to lead to improvements in the treatment of neurodegenerative diseases and stem cell-based replacement therapies. Here, we review the roles played by Wnts in neural stem cells in the developing mouse brain, at neurogenic sites of the adult mouse and in neural stem cell culture models.
Collapse
Affiliation(s)
- Nora Bengoa-Vergniory
- Cell Biology and Stem Cells Unit, CIC bioGUNE, Bilbao, Spain.
- Department of Physiology, Anatomy and Genetics, Oxford University, Oxford, UK.
| | - Robert M Kypta
- Cell Biology and Stem Cells Unit, CIC bioGUNE, Bilbao, Spain.
- Department of Surgery and Cancer, Imperial College London, London, UK.
| |
Collapse
|
70
|
Shahbazi MN, Perez-Moreno M. Connections between cadherin-catenin proteins, spindle misorientation, and cancer. Tissue Barriers 2015; 3:e1045684. [PMID: 26451345 DOI: 10.1080/21688370.2015.1045684] [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] [Received: 02/19/2015] [Revised: 04/18/2015] [Accepted: 04/21/2015] [Indexed: 10/25/2022] Open
Abstract
Cadherin-catenin mediated adhesion is an important determinant of tissue architecture in multicellular organisms. Cancer progression and maintenance is frequently associated with loss of their expression or functional activity, which not only leads to decreased cell-cell adhesion, but also to enhanced tumor cell proliferation and loss of differentiated characteristics. This review is focused on the emerging implications of cadherin-catenin proteins in the regulation of polarized divisions through their connections with the centrosomes, cytoskeleton, tissue tension and signaling pathways; and illustrates how alterations in cadherin-catenin levels or functional activity may render cells susceptible to transformation through the loss of their proliferation-differentiation balance.
Collapse
Affiliation(s)
- Marta N Shahbazi
- Department of Physiology, Development, and Neuroscience; University of Cambridge ; Cambridge, UK
| | - Mirna Perez-Moreno
- Epithelial Cell Biology Group; Cancer Cell Biology Program; Spanish National Cancer Research Centre ; Madrid, Spain
| |
Collapse
|
71
|
Cui X, Pertile R, Liu P, Eyles DW. Vitamin D regulates tyrosine hydroxylase expression: N-cadherin a possible mediator. Neuroscience 2015; 304:90-100. [PMID: 26210580 DOI: 10.1016/j.neuroscience.2015.07.048] [Citation(s) in RCA: 84] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2015] [Revised: 07/14/2015] [Accepted: 07/14/2015] [Indexed: 10/23/2022]
Abstract
Vitamin D is a neuroactive steroid. Its genomic actions are mediated via the active form of vitamin D, 1,25(OH)2D3, binding to the vitamin D receptor (VDR). The VDR emerges in the rat mesencephalon at embryonic day 12, representing the peak period of dopaminergic cell birth. Our prior studies reveal that developmental vitamin D (DVD)-deficiency alters the ontogeny of dopaminergic neurons in the developing mesencephalon. There is also consistent evidence from others that 1,25(OH)2D3 promotes the survival of dopaminergic neurons in models of dopaminergic toxicity. In both developmental and toxicological studies it has been proposed that 1,25(OH)2D3 may modulate the differentiation and maturation of dopaminergic neurons; however, to date there is lack of direct evidence. The aim of the current study is to investigate this both in vitro using a human SH-SY5Y cell line transfected with rodent VDR and in vivo using a DVD-deficient model. Here we show that in VDR-expressing SH-SY5Y cells, 1,25(OH)2D3 significantly increased production of tyrosine hydroxylase (TH), the rate-limiting enzyme in dopamine synthesis. This effect was dose- and time-dependent, but was not due to an increase in TH-positive cell number, nor was it due to the production of trophic survival factors for dopamine neurons such as glial-derived neurotrophic factor (GDNF). In accordance with 1,25(OH)2D3's anti-proliferative actions in the brain, 1,25(OH)2D3 reduced the percentage of dividing cells from approximately 15-10%. Given the recently reported role of N-cadherin in the direct differentiation of dopaminergic neurons, we examined here whether it may be elevated by 1,25(OH)2D3. We confirmed this in vitro and more importantly, we showed DVD-deficiency decreases N-cadherin expression in the embryonic mesencephalon. In summary, in our in vitro model we have shown 1,25(OH)2D3 increases TH expression, decreases proliferation and elevates N-cadherin, a potential factor that mediates these processes. Accordingly all of these findings are reversed in the developing brain in our DVD-deficiency model. Remarkably our findings in the DVD-deficiency model phenocopy those found in a recent model where N-cadherin was regionally ablated from the mesencephalon. This study has, for the first time, shown that vitamin D directly modulates TH expression and strongly suggests N-cadherin may be a plausible mediator of this process both in vitro and in vivo. Our findings may help to explain epidemiological data linking DVD deficiency with schizophrenia.
Collapse
Affiliation(s)
- X Cui
- Queensland Brain Institute, University of Queensland, Qld 4072, Australia
| | - R Pertile
- Queensland Brain Institute, University of Queensland, Qld 4072, Australia
| | - P Liu
- Queensland Brain Institute, University of Queensland, Qld 4072, Australia
| | - D W Eyles
- Queensland Brain Institute, University of Queensland, Qld 4072, Australia; Queensland Centre for Mental Health Research, Wacol, Qld 4076, Australia.
| |
Collapse
|
72
|
Draganova K, Zemke M, Zurkirchen L, Valenta T, Cantù C, Okoniewski M, Schmid MT, Hoffmans R, Götz M, Basler K, Sommer L. Wnt/β-catenin signaling regulates sequential fate decisions of murine cortical precursor cells. Stem Cells 2015; 33:170-82. [PMID: 25182747 DOI: 10.1002/stem.1820] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2014] [Accepted: 07/30/2014] [Indexed: 12/21/2022]
Abstract
The fate of neural progenitor cells (NPCs) is determined by a complex interplay of intrinsic programs and extrinsic signals, very few of which are known. β-Catenin transduces extracellular Wnt signals, but also maintains adherens junctions integrity. Here, we identify for the first time the contribution of β-catenin transcriptional activity as opposed to its adhesion role in the development of the cerebral cortex by combining a novel β-catenin mutant allele with conditional inactivation approaches. Wnt/β-catenin signaling ablation leads to premature NPC differentiation, but, in addition, to a change in progenitor cell cycle kinetics and an increase in basally dividing progenitors. Interestingly, Wnt/β-catenin signaling affects the sequential fate switch of progenitors, leading to a shortened neurogenic period with decreased number of both deep and upper-layer neurons and later, to precocious astrogenesis. Indeed, a genome-wide analysis highlighted the premature activation of a corticogenesis differentiation program in the Wnt/β-catenin signaling-ablated cortex. Thus, β-catenin signaling controls the expression of a set of genes that appear to act downstream of canonical Wnt signaling to regulate the stage-specific production of appropriate progenitor numbers, neuronal subpopulations, and astroglia in the forebrain.
Collapse
|
73
|
Lai M, Guo Y, Ma J, Yu H, Zhao D, Fan W, Ju X, Sheikh MA, Malik YS, Xiong W, Guo W, Zhu X. Myosin X regulates neuronal radial migration through interacting with N-cadherin. Front Cell Neurosci 2015; 9:326. [PMID: 26347613 PMCID: PMC4539528 DOI: 10.3389/fncel.2015.00326] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2015] [Accepted: 08/06/2015] [Indexed: 11/23/2022] Open
Abstract
Proper brain function depends on correct neuronal migration during development, which is known to be regulated by cytoskeletal dynamics and cell-cell adhesion. Myosin X (Myo10), an uncharacteristic member of the myosin family, is an important regulator of cytoskeleton that modulates cell motilities in many different cellular contexts. We previously reported that Myo10 was required for neuronal migration in the developing cerebral cortex, but the underlying mechanism was still largely unknown. Here, we found that knockdown of Myo10 expression disturbed the adherence of migrating neurons to radial glial fibers through abolishing surface Neuronal cadherin (N-cadherin) expression, thereby impaired neuronal migration in the developmental cortex. Next, we found Myo10 interacted with N-cadherin cellular domain through its FERM domain. Furthermore, we found knockdown of Myo10 disrupted N-cadherin subcellular distribution and led to localization of N-cadherin into Golgi apparatus and endosomal sorting vesicle. Taking together, these results reveal a novel mechanism of Myo10 interacting with N-cadherin and regulating its cell-surface expression, which is required for neuronal adhesion and migration.
Collapse
Affiliation(s)
- Mingming Lai
- Key Laboratory of Molecular Epigenetics, Ministry of Education and Institute of Cytology and Genetics, Northeast Normal University Changchun, China ; Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Dali University Dali, China
| | - Ye Guo
- Key Laboratory of Molecular Epigenetics, Ministry of Education and Institute of Cytology and Genetics, Northeast Normal University Changchun, China
| | - Jun Ma
- Key Laboratory of Molecular Epigenetics, Ministry of Education and Institute of Cytology and Genetics, Northeast Normal University Changchun, China
| | - Huali Yu
- Key Laboratory of Molecular Epigenetics, Ministry of Education and Institute of Cytology and Genetics, Northeast Normal University Changchun, China
| | - Dongdong Zhao
- Key Laboratory of Molecular Epigenetics, Ministry of Education and Institute of Cytology and Genetics, Northeast Normal University Changchun, China
| | - Wenqiang Fan
- Key Laboratory of Molecular Epigenetics, Ministry of Education and Institute of Cytology and Genetics, Northeast Normal University Changchun, China
| | - Xingda Ju
- Key Laboratory of Molecular Epigenetics, Ministry of Education and Institute of Cytology and Genetics, Northeast Normal University Changchun, China
| | - Muhammad A Sheikh
- Key Laboratory of Molecular Epigenetics, Ministry of Education and Institute of Cytology and Genetics, Northeast Normal University Changchun, China
| | - Yousra S Malik
- Key Laboratory of Molecular Epigenetics, Ministry of Education and Institute of Cytology and Genetics, Northeast Normal University Changchun, China
| | - Wencheng Xiong
- Department of Neurology, Georgia Regents University, Augusta GA, USA
| | - Weixiang Guo
- State Key Laboratory for Molecular and Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences Beijing, China
| | - Xiaojuan Zhu
- Key Laboratory of Molecular Epigenetics, Ministry of Education and Institute of Cytology and Genetics, Northeast Normal University Changchun, China ; State Key Laboratory for Molecular and Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences Beijing, China
| |
Collapse
|
74
|
Haque A, Adnan N, Motazedian A, Akter F, Hossain S, Kutsuzawa K, Nag K, Kobatake E, Akaike T. An Engineered N-Cadherin Substrate for Differentiation, Survival, and Selection of Pluripotent Stem Cell-Derived Neural Progenitors. PLoS One 2015; 10:e0135170. [PMID: 26244942 PMCID: PMC4526632 DOI: 10.1371/journal.pone.0135170] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2015] [Accepted: 07/18/2015] [Indexed: 11/18/2022] Open
Abstract
For stem cell-based treatment of neurodegenerative diseases a better understanding of key developmental signaling pathways and robust techniques for producing neurons with highest homogeneity are required. In this study, we demonstrate a method using N-cadherin-based biomimetic substrate to promote the differentiation of mouse embryonic stem cell (ESC)- and induced pluripotent stem cell (iPSC)-derived neural progenitor cells (NPCs) without exogenous neuro-inductive signals. We showed that substrate-dependent activation of N-cadherin reduces Rho/ROCK activation and β-catenin expression, leading to the stimulation of neurite outgrowth and conversion into cells expressing neural/glial markers. Besides, plating dissociated cells on N-cadherin substrate can significantly increase the differentiation yield via suppression of dissociation-induced Rho/ROCK-mediated apoptosis. Because undifferentiated ESCs and iPSCs have low affinity to N-cadherin, plating dissociated cells on N-cadherin-coated substrate increase the homogeneity of differentiation by purging ESCs and iPSCs (~30%) from a mixture of undifferentiated cells with NPCs. Using this label-free cell selection approach we enriched differentiated NPCs plated as monolayer without ROCK inhibitor. Therefore, N-cadherin biomimetic substrate provide a powerful tool for basic study of cell—material interaction in a spatially defined and substrate-dependent manner. Collectively, our approach is efficient, robust and cost effective to produce large quantities of differentiated cells with highest homogeneity and applicable to use with other types of cells.
Collapse
Affiliation(s)
- Amranul Haque
- Department of Biomolecular Engineering, Tokyo Institute of Technology, Yokohama, Japan
| | - Nihad Adnan
- Department of Biomolecular Engineering, Tokyo Institute of Technology, Yokohama, Japan
| | - Ali Motazedian
- Department of Biomolecular Engineering, Tokyo Institute of Technology, Yokohama, Japan
| | - Farhima Akter
- Department of Biological Information, Tokyo Institute of Technology, Yokohama, Japan
| | - Sharif Hossain
- Department of Biomolecular Engineering, Tokyo Institute of Technology, Yokohama, Japan
| | - Koichi Kutsuzawa
- Department of Biomolecular Engineering, Tokyo Institute of Technology, Yokohama, Japan
| | - Kakon Nag
- Department of Biomolecular Engineering, Tokyo Institute of Technology, Yokohama, Japan
| | - Eiry Kobatake
- Department of Biological Information, Tokyo Institute of Technology, Yokohama, Japan
| | - Toshihiro Akaike
- Department of Biomolecular Engineering, Tokyo Institute of Technology, Yokohama, Japan
- * E-mail:
| |
Collapse
|
75
|
Coste C, Neirinckx V, Gothot A, Wislet S, Rogister B. Are neural crest stem cells the missing link between hematopoietic and neurogenic niches? Front Cell Neurosci 2015; 9:218. [PMID: 26136659 PMCID: PMC4469833 DOI: 10.3389/fncel.2015.00218] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2015] [Accepted: 05/22/2015] [Indexed: 12/24/2022] Open
Abstract
Hematopoietic niches are defined as cellular and molecular microenvironments that regulate hematopoietic stem cell (HSC) function together with stem cell autonomous mechanisms. Many different cell types have been characterized as contributors to the formation of HSC niches, such as osteoblasts, endothelial cells, Schwann cells, and mesenchymal progenitors. These mesenchymal progenitors have themselves been classified as CXC chemokine ligand (CXCL) 12-abundant reticular (CAR) cells, stem cell factor expressing cells, or nestin-positive mesenchymal stem cells (MSCs), which have been recently identified as neural crest-derived cells (NCSCs). Together, these cells are spatially associated with HSCs and believed to provide appropriate microenvironments for HSC self-renewal, differentiation, mobilization and hibernation both by cell-cell contact and soluble factors. Interestingly, it appears that regulatory pathways governing the hematopoietic niche homeostasis are operating in the neurogenic niche as well. Therefore, this review paper aims to compare both the regulation of hematopoietic and neurogenic niches, in order to highlight the role of NCSCs and nervous system components in the development and the regulation of the hematopoietic system.
Collapse
Affiliation(s)
- Cécile Coste
- Groupe Interdisciplinaire de Génoprotéomique Appliquée-Neurosciences, Unit of Nervous System Disorders and Treatment, University of Liège Liège, Belgium
| | - Virginie Neirinckx
- Groupe Interdisciplinaire de Génoprotéomique Appliquée-Neurosciences, Unit of Nervous System Disorders and Treatment, University of Liège Liège, Belgium
| | - André Gothot
- Groupe Interdisciplinaire de Génoprotéomique Appliquée-Cardiovascular Sciences, University of Liège Liège, Belgium ; Hematology Department, University Hospital Liège, Belgium
| | - Sabine Wislet
- Groupe Interdisciplinaire de Génoprotéomique Appliquée-Neurosciences, Unit of Nervous System Disorders and Treatment, University of Liège Liège, Belgium
| | - Bernard Rogister
- Groupe Interdisciplinaire de Génoprotéomique Appliquée-Neurosciences, Unit of Nervous System Disorders and Treatment, University of Liège Liège, Belgium ; Groupe Interdisciplinaire de Génoprotéomique Appliquée-Development, Stem Cells and Regenerative Medicine, University of Liège Liège, Belgium ; Neurology Department, University Hospital Liège, Belgium
| |
Collapse
|
76
|
Maki T, Maeda M, Uemura M, Lo EK, Terasaki Y, Liang AC, Shindo A, Choi YK, Taguchi A, Matsuyama T, Takahashi R, Ihara M, Arai K. Potential interactions between pericytes and oligodendrocyte precursor cells in perivascular regions of cerebral white matter. Neurosci Lett 2015; 597:164-9. [PMID: 25936593 DOI: 10.1016/j.neulet.2015.04.047] [Citation(s) in RCA: 81] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2015] [Revised: 04/25/2015] [Accepted: 04/27/2015] [Indexed: 12/16/2022]
Abstract
Pericytes are embedded within basal lamina and play multiple roles in the perivascular niche in brain. Recently, oligodendrocyte precursor cells (OPCs) have also been reported to associate with cerebral endothelium. Is it possible that within this gliovascular locus, there may also exist potential spatial and functional interactions between pericytes and OPCs? Here, we demonstrated that in the perivascular region of cerebral white matter, pericytes and OPCs may attach and support each other. Immunostaining showed that pericytes and OPCs are localized in close contact with each other in mouse white matter at postnatal days 0, 60 and 240. Electron microscopic analysis confirmed that pericytes attached to OPCs via basal lamina in the perivascular region. The close proximity between these two cell types was also observed in postmortem human brains. Functional interaction between pericytes and OPCs was assessed by in vitro media transfer experiments. When OPC cultures were treated with pericyte-conditioned media, OPC number increased. Similarly, pericyte number increased when pericytes were maintained in OPC-conditioned media. Taken together, our data suggest a potential anatomical and functional interaction between pericytes and OPCs in cerebral white matter.
Collapse
Affiliation(s)
- Takakuni Maki
- Neuroprotection Research Laboratory, Departments of Radiology and Neurology, Massachusetts General Hospital and Harvard Medical School, USA
| | - Mitsuyo Maeda
- Department of Regenerative Medicine, Institute of Biomedical Research and Innovation, Kobe, Japan
| | - Maiko Uemura
- Department of Neurology, Graduate School of Medicine, Kyoto University, Japan
| | - Evan K Lo
- Neuroprotection Research Laboratory, Departments of Radiology and Neurology, Massachusetts General Hospital and Harvard Medical School, USA
| | - Yasukazu Terasaki
- Neuroprotection Research Laboratory, Departments of Radiology and Neurology, Massachusetts General Hospital and Harvard Medical School, USA
| | - Anna C Liang
- Neuroprotection Research Laboratory, Departments of Radiology and Neurology, Massachusetts General Hospital and Harvard Medical School, USA
| | - Akihiro Shindo
- Neuroprotection Research Laboratory, Departments of Radiology and Neurology, Massachusetts General Hospital and Harvard Medical School, USA
| | - Yoon Kyung Choi
- Neuroprotection Research Laboratory, Departments of Radiology and Neurology, Massachusetts General Hospital and Harvard Medical School, USA
| | - Akihiko Taguchi
- Department of Regenerative Medicine, Institute of Biomedical Research and Innovation, Kobe, Japan
| | - Tomohiro Matsuyama
- Laboratory of Neurogenesis and CNS Repair, Institute for Advanced Medical Science, Hyogo College of Medicine, Hyogo, Japan
| | - Ryosuke Takahashi
- Department of Neurology, Graduate School of Medicine, Kyoto University, Japan
| | - Masafumi Ihara
- Department of Stroke and Cerebrovascular Diseases, National Cerebral and Cardiovascular Center, Osaka, Japan
| | - Ken Arai
- Neuroprotection Research Laboratory, Departments of Radiology and Neurology, Massachusetts General Hospital and Harvard Medical School, USA.
| |
Collapse
|
77
|
Miyamoto Y, Sakane F, Hashimoto K. N-cadherin-based adherens junction regulates the maintenance, proliferation, and differentiation of neural progenitor cells during development. Cell Adh Migr 2015; 9:183-92. [PMID: 25869655 DOI: 10.1080/19336918.2015.1005466] [Citation(s) in RCA: 76] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
This review addresses our current understanding of the regulatory mechanism by which N-cadherin, a classical cadherin, affects neural progenitor cells (NPCs) during development. N-cadherin is responsible for the integrity of adherens junctions (AJs), which develop in the sub-apical region of NPCs in the neural tube and brain cortex. The apical domain, which contains the sub-apical region, is involved in the switching from symmetric proliferative division to asymmetric neurogenic division of NPCs. In addition, N-cadherin-based AJ is deeply involved in the apico-basal polarity of NPCs and the regulation of Wnt-β-catenin, hedgehog (Hh), and Notch signaling. In this review, we discuss the roles of N-cadherin in the maintenance, proliferation, and differentiation of NPCs through components of AJ, β-catenin and αE-catenin.
Collapse
Key Words
- AJ, adherens junction
- EC, extracellular
- Fox, forkhead box
- Frz, frizzled
- GFAP, glial fibrillary acidic protein
- GSK3β, glycogen synthase kinase 3β
- Hes, hairly/enhancer of split
- Hh, hedgehog
- IP, intermediate progenitor
- KO, knockout
- LEF, lymphocyte enhancer factor
- N-cadherin
- NPC, neural progenitor cell
- Par, partition defective complex protein
- Ptc, Pached
- Smo, smoothened
- Sox2, sry (sex determining region Y)-box containing gene 2
- TA cell, transient amplifying cell; ZO-1, Zonula Occludens-1.
- TCF, T-cell factor
- aPKC, atypical protein kinase C
- adherens junction
- apico-basal polarity
- iPSC, induced pluripotent stem cell
- neural progenitor cells
- ngn2, neurogenin 2
- shRNA, short hairpin RNA
- β-catenin
Collapse
Affiliation(s)
- Yasunori Miyamoto
- a The Graduate School of Humanities and Sciences; Ochanomizu University ; Tokyo , Japan
| | | | | |
Collapse
|
78
|
Deleyrolle L, Sabourin JC, Rothhut B, Fujita H, Guichet PO, Teigell M, Ripoll C, Chauvet N, Perrin F, Mamaeva D, Noda T, Mori K, Yoshihara Y, Hugnot JP. OCAM regulates embryonic spinal cord stem cell proliferation by modulating ErbB2 receptor. PLoS One 2015; 10:e0122337. [PMID: 25875008 PMCID: PMC4395419 DOI: 10.1371/journal.pone.0122337] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2014] [Accepted: 02/10/2015] [Indexed: 01/07/2023] Open
Abstract
The proliferation and differentiation of neural stem cells are tightly controlled by intrinsic and extrinsic cues. Cell adhesion molecules are increasingly recognized as regulators of these processes. Here we report the expression of the olfactory cell adhesion molecule (OCAM/NCAM2/RNCAM) during mouse spinal cord development and in neural stem cells cultured as neurospheres. OCAM is also weakly expressed in the dormant adult stem cell niche around the central canal and is overexpressed after spinal cord injury. Both transmembrane (TM) and glycosylphosphatidylinositol (GPI)-linked isoforms are present in neurospheres. Electron microscopy and internalisation experiments revealed a dynamic trafficking of OCAM between the membrane and intracellular compartments. After differentiation, OCAM remains in neurons and oligodendrocytes whereas no expression is detected in astrocytes. Using OCAM knockout (KO) mice, we found that mutant spinal cord stem cells showed an increased proliferation and self-renewal rates although no effect on differentiation was observed. This effect was reversed by lentivirus-mediated re-introduction of OCAM. Mechanistically, we identified the ErbB2/Neu/HER2 protein as being implicated in the enhanced proliferation of mutant cells. ErbB2 protein expression and phosphorylation level were significantly increased in KO cells whereas no difference was observed at the mRNA level. Overexpression of ErbB2 in wild-type and mutant cells also increased their growth while reintroduction of OCAM in mutant cells reduced the level of phosphorylated ErbB2. These results indicate that OCAM exerts a posttranscriptional control on the ErbB2 signalling in spinal cord stem cells. This study adds further support for considering cell adhesion molecules as regulators of the ErbB signalling.
Collapse
Affiliation(s)
- Loïc Deleyrolle
- Department of Neurosurgery, College of Medicine, University of Florida Gainesville, Gainesville, Florida, United States of America
| | | | - Bernard Rothhut
- INSERM U1051, Institute for Neuroscience, Hôpital Saint Eloi, Montpellier, France
- * E-mail:
| | | | | | - Marisa Teigell
- INSERM U1051, Institute for Neuroscience, Hôpital Saint Eloi, Montpellier, France
| | - Chantal Ripoll
- INSERM U1051, Institute for Neuroscience, Hôpital Saint Eloi, Montpellier, France
| | - Norbert Chauvet
- INSERM U661, Department of Endocrinology, Institute of Functional Genomics, Montpellier, France
- University of Montpellier 2, Montpellier, France
| | - Florence Perrin
- INSERM U1051, Institute for Neuroscience, Hôpital Saint Eloi, Montpellier, France
| | - Daria Mamaeva
- INSERM U1051, Institute for Neuroscience, Hôpital Saint Eloi, Montpellier, France
| | - Tetsuo Noda
- Department of Cell Biology, Cancer Institute, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Kensaku Mori
- Department of Physiology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | | | - Jean-Philippe Hugnot
- INSERM U1051, Institute for Neuroscience, Hôpital Saint Eloi, Montpellier, France
- University of Montpellier 2, Montpellier, France
| |
Collapse
|
79
|
Winter J. MicroRNAs of the miR379-410 cluster: New players in embryonic neurogenesis and regulators of neuronal function. NEUROGENESIS 2015; 2:e1004970. [PMID: 27504472 PMCID: PMC4973610 DOI: 10.1080/23262133.2015.1004970] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/29/2014] [Revised: 09/26/2014] [Accepted: 01/03/2015] [Indexed: 01/29/2023]
Abstract
The imprinted miR379–410 cluster contains 38 microRNAs (miRNAs) that are involved in diverse neurodevelopmental processes and are important regulators of neuronal function. The implications of these miRNAs in neurological diseases have been recently recognized.In the present minireview, the current findings regarding the brain-specific functions of miR379–410 cluster miRNAs are summarized and discussed.
Collapse
Affiliation(s)
- Jennifer Winter
- Institute of Human Genetics; University Medical Centre Mainz ; Mainz, Germany
| |
Collapse
|
80
|
Greenman R, Gorelik A, Sapir T, Baumgart J, Zamor V, Segal-Salto M, Levin-Zaidman S, Aidinis V, Aoki J, Nitsch R, Vogt J, Reiner O. Non-cell autonomous and non-catalytic activities of ATX in the developing brain. Front Neurosci 2015; 9:53. [PMID: 25788872 PMCID: PMC4349085 DOI: 10.3389/fnins.2015.00053] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2014] [Accepted: 02/06/2015] [Indexed: 12/20/2022] Open
Abstract
The intricate formation of the cerebral cortex requires a well-coordinated series of events, which are regulated at the level of cell-autonomous and non-cell autonomous mechanisms. Whereas cell-autonomous mechanisms that regulate cortical development are well-studied, the non-cell autonomous mechanisms remain poorly understood. A non-biased screen allowed us to identify Autotaxin (ATX) as a non-cell autonomous regulator of neural stem cells. ATX (also known as ENPP2) is best known to catalyze lysophosphatidic acid (LPA) production. Our results demonstrate that ATX affects the localization and adhesion of neuronal progenitors in a cell autonomous and non-cell autonomous manner, and strikingly, this activity is independent from its catalytic activity in producing LPA.
Collapse
Affiliation(s)
- Raanan Greenman
- Department of Molecular Genetics, Weizmann Institute of Science Rehovot, Israel
| | - Anna Gorelik
- Department of Molecular Genetics, Weizmann Institute of Science Rehovot, Israel
| | - Tamar Sapir
- Department of Molecular Genetics, Weizmann Institute of Science Rehovot, Israel
| | - Jan Baumgart
- University Medical Center, Institute for Microscopic Anatomy and Neurobiology, Johannes Gutenberg-University Mainz Mainz, Germany ; Central Laboratory Animal Facility, University Medical Center, Johannes Gutenberg-University Mainz Mainz, Germany
| | - Vanessa Zamor
- Department of Molecular Genetics, Weizmann Institute of Science Rehovot, Israel
| | - Michal Segal-Salto
- Department of Molecular Genetics, Weizmann Institute of Science Rehovot, Israel
| | - Smadar Levin-Zaidman
- Department of Chemical Research Support, Weizmann Institute of Science Rehovot, Israel
| | - Vassilis Aidinis
- Division of Immunology, Biomedical Sciences Research Center 'Alexander Fleming' Athens, Greece
| | - Junken Aoki
- Graduate School of Pharmaceutical Sciences, Tohoku University Miyagi, Japan
| | - Robert Nitsch
- University Medical Center, Institute for Microscopic Anatomy and Neurobiology, Johannes Gutenberg-University Mainz Mainz, Germany
| | - Johannes Vogt
- University Medical Center, Institute for Microscopic Anatomy and Neurobiology, Johannes Gutenberg-University Mainz Mainz, Germany
| | - Orly Reiner
- Department of Molecular Genetics, Weizmann Institute of Science Rehovot, Israel
| |
Collapse
|
81
|
Abstract
The arrival of multicellularity in evolution facilitated cell-cell signaling in conjunction with adhesion. As the ectodomains of cadherins interact with each other directly in trans (as well as in cis), spanning the plasma membrane and associating with multiple other entities, cadherins enable the transduction of "outside-in" or "inside-out" signals. We focus this review on signals that originate from the larger family of cadherins that are inwardly directed to the nucleus, and thus have roles in gene control or nuclear structure-function. The nature of cadherin complexes varies considerably depending on the type of cadherin and its context, and we will address some of these variables for classical cadherins versus other family members. Substantial but still fragmentary progress has been made in understanding the signaling mediators used by varied cadherin complexes to coordinate the state of cell-cell adhesion with gene expression. Evidence that cadherin intracellular binding partners also localize to the nucleus is a major point of interest. In some models, catenins show reduced binding to cadherin cytoplasmic tails favoring their engagement in gene control. When bound, cadherins may serve as stoichiometric competitors of nuclear signals. Cadherins also directly or indirectly affect numerous signaling pathways (e.g., Wnt, receptor tyrosine kinase, Hippo, NFκB, and JAK/STAT), enabling cell-cell contacts to touch upon multiple biological outcomes in embryonic development and tissue homeostasis.
Collapse
Affiliation(s)
- Pierre D McCrea
- Department of Genetics, University of Texas MD Anderson Cancer Center; Program in Genes & Development, Graduate School in Biomedical Sciences, Houston, Texas, USA.
| | - Meghan T Maher
- Department of Biology, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Cara J Gottardi
- Cellular and Molecular Biology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA; Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA.
| |
Collapse
|
82
|
Banda E, McKinsey A, Germain N, Carter J, Anderson NC, Grabel L. Cell polarity and neurogenesis in embryonic stem cell-derived neural rosettes. Stem Cells Dev 2015; 24:1022-33. [PMID: 25472739 DOI: 10.1089/scd.2014.0415] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Embryonic stem cells (ESCs) undergoing neural differentiation form radial arrays of neural stem cells, termed neural rosettes. These structures manifest many of the properties associated with embryonic and adult neurogenesis, including cell polarization, interkinetic nuclear migration (INM), and a gradient of neuronal differentiation. We now identify novel rosette structural features that serve to localize key regulators of neurogenesis. Cells within neural rosettes have specialized basal as well as apical surfaces, based on localization of the extracellular matrix receptor β1 integrin. Apical processes of cells in mature rosettes terminate at the lumen, where adherens junctions are apparent. Primary cilia are randomly distributed in immature rosettes and tightly associated with the neural stem cell's apical domain as rosettes mature. Components of two signaling pathways known to regulate neurogenesis in vivo and in rosettes, Hedgehog and Notch, are apically localized, with the Hedgehog effector Smoothened (Smo) associated with primary cilia and the Notch pathway γ-secretase subunit Presenilin 2 associated with the adherens junction. Increased neuron production upon treatment with the Notch inhibitor DAPT suggests a major role for Notch signaling in maintaining the neural stem cell state, as previously described. A less robust outcome was observed with manipulation of Hedgehog levels, though consistent with a role in neural stem cell survival or proliferation. Inhibition of both pathways resulted in an additive effect. These data support a model by which cells extending a process to the rosette lumen maintain neural stem cell identity whereas release from this association, either through asymmetric cell division or apical abscission, promotes neuronal differentiation.
Collapse
Affiliation(s)
- Erin Banda
- 1 Biology Department, Wesleyan University , Middletown, Connecticut
| | | | | | | | | | | |
Collapse
|
83
|
Cdo suppresses canonical Wnt signalling via interaction with Lrp6 thereby promoting neuronal differentiation. Nat Commun 2014; 5:5455. [PMID: 25406935 DOI: 10.1038/ncomms6455] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2014] [Accepted: 10/02/2014] [Indexed: 01/23/2023] Open
Abstract
Canonical Wnt signalling regulates expansion of neural progenitors and functions as a dorsalizing signal in the developing forebrain. In contrast, the multifunctional co-receptor Cdo promotes neuronal differentiation and is important for the function of the ventralizing signal, Shh. Here we show that Cdo negatively regulates Wnt signalling during neurogenesis. Wnt signalling is enhanced in Cdo-deficient cells, leading to impaired neuronal differentiation. The ectodomains of Cdo and Lrp6 interact via the Ig2 repeat of Cdo and the LDLR repeats of Lrp6, and the Cdo Ig2 repeat is necessary for Cdo-dependent Wnt inhibition. Furthermore, the Cdo-deficient dorsal forebrain displays stronger Wnt signalling activity, increased cell proliferation and enhanced expression of the dorsal markers and Wnt targets, Pax6, Gli3, Axin2. Therefore, in addition to promoting ventral central nervous system cell fates with Shh, Cdo promotes neuronal differentiation by suppression of Wnt signalling and provides a direct link between two major dorsoventral morphogenetic signalling pathways.
Collapse
|
84
|
Cherry JF, Bennett NK, Schachner M, Moghe PV. Engineered N-cadherin and L1 biomimetic substrates concertedly promote neuronal differentiation, neurite extension and neuroprotection of human neural stem cells. Acta Biomater 2014; 10:4113-26. [PMID: 24914828 DOI: 10.1016/j.actbio.2014.06.001] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2014] [Revised: 04/26/2014] [Accepted: 06/01/2014] [Indexed: 02/05/2023]
Abstract
We investigated the design of neurotrophic biomaterial constructs for human neural stem cells, guided by neural developmental cues of N-cadherin and L1 adhesion molecules. Polymer substrates fabricated either as two-dimensional (2-D) films or three-dimensional (3-D) microfibrous scaffolds were functionalized with fusion chimeras of N-cadherin-Fc alone and in combination with L1-Fc, and the effects on differentiation, neurite extension and survival of H9 human-embryonic-stem-cell-derived neural stem cells (H9-NSCs) were quantified. Combinations of N-cadherin and L1-Fc co-operatively enhanced neuronal differentiation profiles, indicating the critical nature of the two complementary developmental cues. Notably, substrates presenting low levels of N-cadherin-Fc concentrations, combined with proportionately higher L1-Fc concentration, most enhanced neurite outgrowth and the degree of MAP2+ and neurofilament-M+ H9-NSCs. Low N-cadherin-Fc alone promoted improved cell survival following oxidative stress, compared to higher concentrations of N-cadherin-Fc alone or combinations with L1-Fc. Pharmacological and antibody blockage studies revealed that substrates presenting low levels of N-cadherin are functionally competent so long as they elicit a threshold signal mediated by homophilic N-cadherin and fibroblast growth factor signaling. Overall, these studies highlight the ability of optimal combinations of N-cadherin and L1 to recapitulate a "neurotrophic" microenvironment that enhances human neural stem cell differentiation and neurite outgrowth. Additionally, 3-D fibrous scaffolds presenting low N-cadherin-Fc further enhanced the survival of H9-NSCs compared to equivalent 2-D films. This indicates that similar biofunctionalization approaches based on N-cadherin and L1 can be translated to 3-D "transplantable" scaffolds with enhanced neurotrophic behaviors. Thus, the insights from this study have fundamental and translational impacts for neural-stem-cell-based regenerative medicine.
Collapse
Affiliation(s)
- Jocie F Cherry
- Department of Biomedical Engineering, Rutgers University, 599 Taylor Road, Piscataway, NJ 08854, USA
| | - Neal K Bennett
- Department of Biomedical Engineering, Rutgers University, 599 Taylor Road, Piscataway, NJ 08854, USA
| | - Melitta Schachner
- W.M. Keck Center for Collaborative Neuroscience, Department of Cell Biology and Neuroscience, Rutgers University, 599 Taylor Road, Piscataway, NJ 08854, USA; Center for Neuroscience, Shantou University Medical College, 22 Xin Ling Road, Shantou 515041, People's Republic of China
| | - Prabhas V Moghe
- Department of Biomedical Engineering, Rutgers University, 599 Taylor Road, Piscataway, NJ 08854, USA; Department of Chemical and Biochemical Engineering, Rutgers University, 599 Taylor Road, Piscataway, NJ 08854, USA.
| |
Collapse
|
85
|
Lai YJ, Li MY, Yang CY, Huang KH, Tsai JC, Wang TW. TRIP6 regulates neural stem cell maintenance in the postnatal mammalian subventricular zone. Dev Dyn 2014; 243:1130-42. [DOI: 10.1002/dvdy.24161] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2014] [Revised: 06/25/2014] [Accepted: 06/25/2014] [Indexed: 11/06/2022] Open
Affiliation(s)
- Yun-Ju Lai
- Department of Life Science; National Taiwan Normal University; Taipei Taiwan
| | - Ming-Yang Li
- Department of Life Science; National Taiwan Normal University; Taipei Taiwan
| | - Cheng-Yao Yang
- Department of Life Science; National Taiwan Normal University; Taipei Taiwan
| | - Kao-Hua Huang
- Department of Life Science; National Taiwan Normal University; Taipei Taiwan
| | - Jui-Cheng Tsai
- Department of Life Science; National Taiwan Normal University; Taipei Taiwan
| | - Tsu-Wei Wang
- Department of Life Science; National Taiwan Normal University; Taipei Taiwan
- Brain Research Center; National Yang-Ming University; Taipei Taiwan
| |
Collapse
|
86
|
Klingener M, Chavali M, Singh J, McMillan N, Coomes A, Dempsey PJ, Chen EI, Aguirre A. N-cadherin promotes recruitment and migration of neural progenitor cells from the SVZ neural stem cell niche into demyelinated lesions. J Neurosci 2014; 34:9590-606. [PMID: 25031401 PMCID: PMC4099541 DOI: 10.1523/jneurosci.3699-13.2014] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2013] [Revised: 06/04/2014] [Accepted: 06/06/2014] [Indexed: 01/01/2023] Open
Abstract
Discrete cellular microenvironments regulate stem cell pools and their development, as well as function in maintaining tissue homeostasis. Although the signaling elements modulating neural progenitor cells (NPCs) of the adult subventricular zone (SVZ) niche are fairly well understood, the pathways activated following injury and the resulting outcomes, are less clear. In the present study, we used mouse models of demyelination and proteomics analysis to identify molecular cues present in the adult SVZ niche during injury, and analyzed their role on NPCs in the context of promoting myelin repair. Proteomic analysis of SVZ tissue from mice with experimental demyelination identified several proteins that are known to play roles in NPC proliferation, adhesion, and migration. Among the proteins found to be upregulated were members of the N-cadherin signaling pathway. During the onset of demyelination in the subcortical white matter (SCWM), activation of epidermal growth factor receptor (EGFR) signaling in SVZ NPCs stimulates the interaction between N-cadherin and ADAM10. Upon cleavage and activation of N-cadherin signaling by ADAM10, NPCs undergo cytoskeletal rearrangement and polarization, leading to enhanced migration out of the SVZ into demyelinated lesions of the SCWM. Genetically disrupting either EGFR signaling or ADAM10 inhibits this pathway, preventing N-cadherin regulated NPC polarization and migration. Additionally, in vivo experiments using N-cadherin gain- and loss-of-function approaches demonstrated that N-cadherin enhances the recruitment of SVZ NPCs into demyelinated lesions. Our data revealed that EGFR-dependent N-cadherin signaling physically initiated by ADAM10 cleavage is the response of the SVZ niche to promote repair of the injured brain.
Collapse
Affiliation(s)
- Michael Klingener
- State University of New York at Stony Brook University, Departments of Pharmacological Science and
| | - Manideep Chavali
- State University of New York at Stony Brook University, Departments of Pharmacological Science and Materials Science and Engineering, Stony Brook, New York 11794
| | - Jagdeep Singh
- State University of New York at Stony Brook University, Departments of Pharmacological Science and
| | - Nadia McMillan
- State University of New York at Stony Brook University, Departments of Pharmacological Science and
| | - Alexandra Coomes
- State University of New York at Stony Brook University, Departments of Pharmacological Science and Stony Brook University Proteomics Center, School of Medicine, Stony Brook, New York 11794
| | - Peter J Dempsey
- Department of Pediatrics and Communicable Diseases, University of Michigan, Ann Arbor, Michigan 48109, and
| | - Emily I Chen
- State University of New York at Stony Brook University, Departments of Pharmacological Science and Stony Brook University Proteomics Center, School of Medicine, Stony Brook, New York 11794
| | - Adan Aguirre
- State University of New York at Stony Brook University, Departments of Pharmacological Science and
| |
Collapse
|
87
|
Taverna E, Götz M, Huttner WB. The cell biology of neurogenesis: toward an understanding of the development and evolution of the neocortex. Annu Rev Cell Dev Biol 2014; 30:465-502. [PMID: 25000993 DOI: 10.1146/annurev-cellbio-101011-155801] [Citation(s) in RCA: 540] [Impact Index Per Article: 49.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Neural stem and progenitor cells have a central role in the development and evolution of the mammalian neocortex. In this review, we first provide a set of criteria to classify the various types of cortical stem and progenitor cells. We then discuss the issue of cell polarity, as well as specific subcellular features of these cells that are relevant for their modes of division and daughter cell fate. In addition, cortical stem and progenitor cell behavior is placed into a tissue context, with consideration of extracellular signals and cell-cell interactions. Finally, the differences across species regarding cortical stem and progenitor cells are dissected to gain insight into key developmental and evolutionary mechanisms underlying neocortex expansion.
Collapse
Affiliation(s)
- Elena Taverna
- Max-Planck Institute of Molecular Cell Biology and Genetics, 01307 Dresden, Germany;
| | | | | |
Collapse
|
88
|
Hatakeyama J, Wakamatsu Y, Nagafuchi A, Kageyama R, Shigemoto R, Shimamura K. Cadherin-based adhesions in the apical endfoot are required for active Notch signaling to control neurogenesis in vertebrates. Development 2014; 141:1671-82. [PMID: 24715457 DOI: 10.1242/dev.102988] [Citation(s) in RCA: 70] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
The development of the vertebrate brain requires an exquisite balance between proliferation and differentiation of neural progenitors. Notch signaling plays a pivotal role in regulating this balance, yet the interaction between signaling and receiving cells remains poorly understood. We have found that numerous nascent neurons and/or intermediate neurogenic progenitors expressing the ligand of Notch retain apical endfeet transiently at the ventricular lumen that form adherens junctions (AJs) with the endfeet of progenitors. Forced detachment of the apical endfeet of those differentiating cells by disrupting AJs resulted in precocious neurogenesis that was preceded by the downregulation of Notch signaling. Both Notch1 and its ligand Dll1 are distributed around AJs in the apical endfeet, and these proteins physically interact with ZO-1, a constituent of the AJ. Furthermore, live imaging of a fluorescently tagged Notch1 demonstrated its trafficking from the apical endfoot to the nucleus upon cleavage. Our results identified the apical endfoot as the central site of active Notch signaling to securely prohibit inappropriate differentiation of neural progenitors.
Collapse
Affiliation(s)
- Jun Hatakeyama
- Department of Brain Morphogenesis, Institute of Molecular Embryology and Genetics, Kumamoto University, 2-2-1 Honjo, Chuo-ku, Kumamoto 860-0811, Japan
| | | | | | | | | | | |
Collapse
|
89
|
Trümbach D, Prakash N. The conserved miR-8/miR-200 microRNA family and their role in invertebrate and vertebrate neurogenesis. Cell Tissue Res 2014; 359:161-77. [PMID: 24875007 DOI: 10.1007/s00441-014-1911-z] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2014] [Accepted: 05/05/2014] [Indexed: 01/24/2023]
Abstract
Since their discovery in the early 1990s, microRNAs have emerged as key components of the post-transcriptional regulation of gene expression. MicroRNAs occur in the plant and animal kingdoms, with the numbers of microRNAs encoded in the genome increasing together with the evolutionary expansion of the phyla. By base-pairing with complementary sequences usually located within the 3' untranslated region, microRNAs target mRNAs for degradation, destabilization and/or translational inhibition. Because one microRNA can have many, if not hundreds, of target mRNAs and because one mRNA can, in turn, be targeted by many microRNAs, these small single-stranded RNAs can exert extensive pleiotropic functions during the development, adulthood and ageing of an organism. Specific functions of an increasing number of microRNAs have been described for the invertebrate and vertebrate nervous systems. Among these, the miR-8/miR-200 microRNA family has recently emerged as an important regulator of neurogenesis and gliogenesis and of adult neural homeostasis in the central nervous system of fruit flies, zebrafish and rodents. This highly conserved microRNA family consists of a single ortholog in the fruit fly (miR-8) and five members in vertebrates (miR-200a, miR-200b, miR-200c, miR-141 and miR-429). Here, we review our current knowledge about the functions of the miR-8/miR-200 microRNA family during invertebrate and vertebrate neural development and adult homeostasis and, in particular, about their role in the regulation of neural stem/progenitor cell proliferation, cell cycle exit, transition to a neural precursor/neuroblast state, neuronal differentiation and cell survival and during glial cell growth and differentiation into mature oligodendrocytes.
Collapse
Affiliation(s)
- Dietrich Trümbach
- Institute of Developmental Genetics, Helmholtz Zentrum München, Deutsches Forschungszentrum für Gesundheit und Umwelt, Ingolstädter Landstrasse 1, 85764, Neuherberg, Germany
| | | |
Collapse
|
90
|
Kuwahara A, Sakai H, Xu Y, Itoh Y, Hirabayashi Y, Gotoh Y. Tcf3 represses Wnt-β-catenin signaling and maintains neural stem cell population during neocortical development. PLoS One 2014; 9:e94408. [PMID: 24832538 PMCID: PMC4022625 DOI: 10.1371/journal.pone.0094408] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2013] [Accepted: 03/15/2014] [Indexed: 01/02/2023] Open
Abstract
During mouse neocortical development, the Wnt–β-catenin signaling pathway plays essential roles in various phenomena including neuronal differentiation and proliferation of neural precursor cells (NPCs). Production of the appropriate number of neurons without depletion of the NPC population requires precise regulation of the balance between differentiation and maintenance of NPCs. However, the mechanism that suppresses Wnt signaling to prevent premature neuronal differentiation of NPCs is poorly understood. We now show that the HMG box transcription factor Tcf3 (also known as Tcf7l1) contributes to this mechanism. Tcf3 is highly expressed in undifferentiated NPCs in the mouse neocortex, and its expression is reduced in intermediate neuronal progenitors (INPs) committed to the neuronal fate. We found Tcf3 to be a repressor of Wnt signaling in neocortical NPCs in a reporter gene assay. Tcf3 bound to the promoter of the proneural bHLH gene Neurogenin1 (Neurog1) and repressed its expression. Consistent with this, Tcf3 repressed neuronal differentiation and increased the self-renewal activity of NPCs. We also found that Wnt signal stimulation reduces the level of Tcf3, and increases those of Tcf1 (also known as Tcf7) and Lef1, positive mediators of Wnt signaling, in NPCs. Together, these results suggest that Tcf3 antagonizes Wnt signaling in NPCs, thereby maintaining their undifferentiated state in the neocortex and that Wnt signaling promotes the transition from Tcf3-mediated repression to Tcf1/Lef1-mediated enhancement of Wnt signaling, constituting a positive feedback loop that facilitates neuronal differentiation.
Collapse
Affiliation(s)
- Atsushi Kuwahara
- Laboratory of Cell Signaling, Institute of Molecular and Cellular Biosciences, The University of Tokyo, Tokyo, Japan
| | - Hiroshi Sakai
- Laboratory of Cell Signaling, Institute of Molecular and Cellular Biosciences, The University of Tokyo, Tokyo, Japan
| | - Yuanjiang Xu
- Laboratory of Cell Signaling, Institute of Molecular and Cellular Biosciences, The University of Tokyo, Tokyo, Japan
| | - Yasuhiro Itoh
- Laboratory of Cell Signaling, Institute of Molecular and Cellular Biosciences, The University of Tokyo, Tokyo, Japan
| | - Yusuke Hirabayashi
- Laboratory of Cell Signaling, Institute of Molecular and Cellular Biosciences, The University of Tokyo, Tokyo, Japan
- * E-mail:
| | - Yukiko Gotoh
- Laboratory of Cell Signaling, Institute of Molecular and Cellular Biosciences, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
91
|
Zhang P, Wu C, Liu N, Niu L, Yan Z, Feng Y, Xu R. Protocadherin 11 x regulates differentiation and proliferation of neural stem cell in vitro and in vivo. J Mol Neurosci 2014; 54:199-210. [PMID: 24647733 DOI: 10.1007/s12031-014-0275-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2014] [Accepted: 02/25/2014] [Indexed: 01/11/2023]
Abstract
Protocadherin 11 X-linked (Pcdh11x) protein is a member of the cadherin superfamily with established roles in cell adhesion. Previous studies have shown the molecular biology and possible relevance of Pcdh11x with neurological disease in humans. However, little is known about the neurophysiological function of Pcdh11x in neural development. Here, we verified that Pcdh11x is primarily expressed in various brain areas including the cortex, hippocampus, and ventricular/subventricular zone (VZ/SVZ) at different embryonic stages. Furthermore, both in vitro and in vivo experiments showed that Pcdh11x decreased neural differentiation but increased the neural proliferation. These observations demonstrate a crucial function for Pcdh11x during the development of central nervous system.
Collapse
Affiliation(s)
- Peng Zhang
- Bayi Brain Hospital, Bayi Clinical Medical Institute, Southern Medical University, Beijing, China
| | | | | | | | | | | | | |
Collapse
|
92
|
Rago L, Beattie R, Taylor V, Winter J. miR379-410 cluster miRNAs regulate neurogenesis and neuronal migration by fine-tuning N-cadherin. EMBO J 2014; 33:906-20. [PMID: 24614228 DOI: 10.1002/embj.201386591] [Citation(s) in RCA: 79] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
N-cadherin-mediated adhesion is essential for maintaining the tissue architecture and stem cell niche in the developing neocortex. N-cadherin expression level is precisely and dynamically controlled throughout development; however, the underlying regulatory mechanisms remain largely unknown. MicroRNAs (miRNAs) play an important role in the regulation of protein expression and subcellular localisation. In this study, we show that three miRNAs belonging to the miR379-410 cluster regulate N-cadherin expression levels in neural stem cells and migrating neurons. The overexpression of these three miRNAs in radial glial cells repressed N-cadherin expression and increased neural stem cell differentiation and neuronal migration. This phenotype was rescued when N-cadherin was expressed from a miRNA-insensitive construct. Transient abrogation of the miRNAs reduced stem cell differentiation and increased cell proliferation. The overexpression of these miRNAs specifically in newborn neurons delayed migration into the cortical plate, whereas the knockdown increased migration. Collectively, our results indicate a novel role for miRNAs of the miR379-410 cluster in the fine-tuning of N-cadherin expression level and in the regulation of neurogenesis and neuronal migration in the developing neocortex.
Collapse
Affiliation(s)
- Luciano Rago
- Department of Molecular Embryology, Max Planck Institute of Immunobiology and Epigenetics, Freiburg, Germany
| | | | | | | |
Collapse
|
93
|
Bielen H, Houart C. The Wnt cries many: Wnt regulation of neurogenesis through tissue patterning, proliferation, and asymmetric cell division. Dev Neurobiol 2014; 74:772-80. [DOI: 10.1002/dneu.22168] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2013] [Revised: 11/23/2013] [Accepted: 01/28/2014] [Indexed: 01/09/2023]
Affiliation(s)
- Holger Bielen
- MRC Centre for Developmental Neurobiology; King's College London, Guy's Campus; London SE1 1UL United Kingdom
| | - Corinne Houart
- MRC Centre for Developmental Neurobiology; King's College London, Guy's Campus; London SE1 1UL United Kingdom
| |
Collapse
|
94
|
N-cadherin sustains motility and polarity of future cortical interneurons during tangential migration. J Neurosci 2014; 33:18149-60. [PMID: 24227724 DOI: 10.1523/jneurosci.0593-13.2013] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
In the developing brain, cortical GABAergic interneurons migrate long distances from the medial ganglionic eminence (MGE) in which they are generated, to the cortex in which they settle. MGE cells express the cell adhesion molecule N-cadherin, a homophilic cell-cell adhesion molecule that regulates numerous steps of brain development, from neuroepithelium morphogenesis to synapse formation. N-cadherin is also expressed in embryonic territories crossed by MGE cells during their migration. In this study, we demonstrate that N-cadherin is a key player in the long-distance migration of future cortical interneurons. Using N-cadherin-coated substrate, we show that N-cadherin-dependent adhesion promotes the migration of mouse MGE cells in vitro. Conversely, mouse MGE cells electroporated with a construct interfering with cadherin function show reduced cell motility, leading process instability, and impaired polarization associated with abnormal myosin IIB dynamics. In vivo, the capability of electroporated MGE cells to invade the developing cortical plate is altered. Using genetic ablation of N-cadherin in mouse embryos, we show that N-cadherin-depleted MGEs are severely disorganized. MGE cells hardly exit the disorganized proliferative area. N-cadherin ablation at the postmitotic stage, which does not affect MGE morphogenesis, alters MGE cell motility and directionality. The tangential migration to the cortex of N-cadherin ablated MGE cells is delayed, and their radial migration within the cortical plate is perturbed. Altogether, these results identify N-cadherin as a pivotal adhesion substrate that activates cell motility in future cortical interneurons and maintains cell polarity over their long-distance migration to the developing cortex.
Collapse
|
95
|
Axin directs the amplification and differentiation of intermediate progenitors in the developing cerebral cortex. Neuron 2013; 79:665-79. [PMID: 23972596 DOI: 10.1016/j.neuron.2013.06.017] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/07/2013] [Indexed: 01/07/2023]
Abstract
UNLABELLED The expansion of the mammalian cerebral cortex is safeguarded by a concerted balance between amplification and neuronal differentiation of intermediate progenitors (IPs). Nonetheless, the molecular controls governing these processes remain unclear. We found that the scaffold protein Axin is a critical regulator that determines the IP population size and ultimately the number of neurons during neurogenesis in the developing cerebral cortex. The increase of the IP pool is mediated by the interaction between Axin and GSK-3 in the cytoplasmic compartments of the progenitors. Importantly, as development proceeds, Axin becomes enriched in the nucleus to trigger neuronal differentiation via β-catenin activation. The nuclear localization of Axin and hence the switch of IPs from proliferative to differentiative status are strictly controlled by the Cdk5-dependent phosphorylation of Axin at Thr485. Our results demonstrate an important Axin-dependent regulatory mechanism in neurogenesis, providing potential insights into the evolutionary expansion of the cerebral cortex. VIDEO ABSTRACT
Collapse
|
96
|
Mitotic spindle orientation predicts outer radial glial cell generation in human neocortex. Nat Commun 2013; 4:1665. [PMID: 23575669 PMCID: PMC3625970 DOI: 10.1038/ncomms2647] [Citation(s) in RCA: 167] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2012] [Accepted: 02/25/2013] [Indexed: 12/23/2022] Open
Abstract
The human neocortex is increased in size and complexity as compared with most other species. Neocortical expansion has recently been attributed to protracted neurogenesis by outer radial glial cells in the outer subventricular zone, a region present in humans but not in rodents. The mechanisms of human outer radial glial cell generation are unknown, but are proposed to involve division of ventricular radial glial cells; neural stem cells present in all developing mammals. Here we show that human ventricular radial glial cells produce outer radial glial cells and seed formation of the outer subventricular zone via horizontal divisions, which occur more frequently in humans than in rodents. We further find that outer radial glial cell mitotic behaviour is cell intrinsic, and that the basal fibre, inherited by outer radial glial cells after ventricular radial glial division, determines cleavage angle. Our results suggest that altered regulation of mitotic spindle orientation increased outer radial glial cell number, and ultimately neuronal number, during human brain evolution.
Collapse
|
97
|
SUN YUANJIE, KIM NAMHO, JI LITING, KIM SEUNGHYUK, LEE JONGHO, RHEE HAEJIN. Lysophosphatidic acid activates β-catenin/T cell factor signaling, which contributes to the suppression of apoptosis in H19-7 cells. Mol Med Rep 2013; 8:1729-33. [DOI: 10.3892/mmr.2013.1743] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2013] [Accepted: 10/03/2013] [Indexed: 11/06/2022] Open
|
98
|
Katayama KI, Imai F, Campbell K, Lang RA, Zheng Y, Yoshida Y. RhoA and Cdc42 are required in pre-migratory progenitors of the medial ganglionic eminence ventricular zone for proper cortical interneuron migration. Development 2013; 140:3139-45. [PMID: 23861058 DOI: 10.1242/dev.092585] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Cortical interneurons arise from the ganglionic eminences in the ventral telencephalon and migrate tangentially to the cortex. Although RhoA and Cdc42, members of the Rho family of small GTPases, have been implicated in regulating neuronal migration, their respective roles in the tangential migration of cortical interneurons remain unknown. Here we show that loss of RhoA and Cdc42 in the ventricular zone (VZ) of the medial ganglionic eminence (MGE) using Olig2-Cre mice causes moderate or severe defects in the migration of cortical interneurons, respectively. Furthermore, RhoA- or Cdc42-deleted MGE cells exhibit impaired migration in vitro. To determine whether RhoA and Cdc42 directly regulate the motility of cortical interneurons during migration, we deleted RhoA and Cdc42 in the subventricular zone (SVZ), where more fate-restricted progenitors are located within the ganglionic eminences, using Dlx5/6-Cre-ires-EGFP (Dlx5/6-CIE) mice. Deletion of either gene within the SVZ does not cause any obvious defects in cortical interneuron migration, indicating that cell motility is not dependent upon RhoA or Cdc42. These findings provide genetic evidence that RhoA and Cdc42 are required in progenitors of the MGE in the VZ, but not the SVZ, for proper cortical interneuron migration.
Collapse
Affiliation(s)
- Kei-ichi Katayama
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH 45229, USA.
| | | | | | | | | | | |
Collapse
|
99
|
Itoh Y, Tyssowski K, Gotoh Y. Transcriptional coupling of neuronal fate commitment and the onset of migration. Curr Opin Neurobiol 2013; 23:957-64. [PMID: 23973158 DOI: 10.1016/j.conb.2013.08.003] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2013] [Revised: 08/05/2013] [Accepted: 08/05/2013] [Indexed: 12/18/2022]
Abstract
During mammalian CNS development, when the neural precursor cells commit to the neuronal fate they must delaminate and migrate toward the pial surface in order to reach the appropriate final location. Thus, the coordination of delamination and fate commitment is important in creating the correct structure. Although previous studies have proposed that spindle orientation during mitosis plays a role in both delamination and fate commitment, thus coordinating these events, subsequent studies have challenged this model. Recent work has identified several transcriptional mechanisms associated with neurogenesis that inhibit cell adhesion of newborn neurons and intermediate neuronal progenitors, thereby triggering delamination and linking it with fate commitment.
Collapse
Affiliation(s)
- Yasuhiro Itoh
- Institute of Molecular and Cellular Biosciences, University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo 113-0032, Japan.
| | | | | |
Collapse
|
100
|
de Back W, Zimm R, Brusch L. Transdifferentiation of pancreatic cells by loss of contact-mediated signaling. BMC SYSTEMS BIOLOGY 2013; 7:77. [PMID: 23938152 PMCID: PMC3751562 DOI: 10.1186/1752-0509-7-77] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/10/2012] [Accepted: 07/04/2013] [Indexed: 12/01/2022]
Abstract
Background Replacement of dysfunctional β-cells in the islets of Langerhans by transdifferentiation of pancreatic acinar cells has been proposed as a regenerative therapy for diabetes. Adult acinar cells spontaneously revert to a multipotent state upon tissue dissociation in vitro and can be stimulated to redifferentiate into β-cells. Despite accumulating evidence that contact-mediated signals are involved, the mechanisms regulating acinar-to-islet cell transdifferentiation remain poorly understood. Results In this study, we propose that the crosstalk between two contact-mediated signaling mechanisms, lateral inhibition and lateral stabilization, controls cell fate stability and transdifferentiation of pancreatic cells. Analysis of a mathematical model combining gene regulation with contact-mediated signaling reveals the multistability of acinar and islet cell fates. Inhibition of one or both modes of signaling results in transdifferentiation from the acinar to the islet cell fate, either by dedifferentiation to a multipotent state or by direct lineage switching. Conclusions This study provides a theoretical framework to understand the role of contact-mediated signaling in pancreatic cell fate control that may help to improve acinar-to-islet cell transdifferentiation strategies for β-cell neogenesis.
Collapse
Affiliation(s)
- Walter de Back
- Center for Information Services and High Performance Computing, Technische Universität Dresden, Dresden, 01062, Germany
| | | | | |
Collapse
|