1
|
Saade M, Martí E. Early spinal cord development: from neural tube formation to neurogenesis. Nat Rev Neurosci 2025; 26:195-213. [PMID: 39915695 DOI: 10.1038/s41583-025-00906-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/20/2025] [Indexed: 03/26/2025]
Abstract
As one of the simplest and most evolutionarily conserved parts of the vertebrate nervous system, the spinal cord serves as a key model for understanding the principles of nervous system construction. During embryonic development, the spinal cord originates from a population of bipotent stem cells termed neuromesodermal progenitors, which are organized within a transient embryonic structure known as the neural tube. Neural tube morphogenesis differs along its anterior-to-posterior axis: most of the neural tube (including the regions that will develop into the brain and the anterior spinal cord) forms via the bending and dorsal fusion of the neural groove, but the establishment of the posterior region of the neural tube involves de novo formation of a lumen within a solid medullary cord. The early spinal cord primordium consists of highly polarized neural progenitor cells organized into a pseudostratified epithelium. Tight regulation of the cell division modes of these progenitors drives the embryonic growth of the neural tube and initiates primary neurogenesis. A rich history of observational and functional studies across various vertebrate models has advanced our understanding of the cellular events underlying spinal cord development, and these foundational studies are beginning to inform our knowledge of human spinal cord development.
Collapse
Affiliation(s)
- Murielle Saade
- Department of Cells and Tissues, Instituto de Biología Molecular de Barcelona CSIC, Barcelona, Spain.
| | - Elisa Martí
- Department of Cells and Tissues, Instituto de Biología Molecular de Barcelona CSIC, Barcelona, Spain.
| |
Collapse
|
2
|
Cho SH, Kim JH, Kim S. Perturbed cell cycle phase-dependent positioning and nuclear migration of retinal progenitors along the apico-basal axis underlie global retinal disorganization in the LCA8-like mouse model. Dev Biol 2025; 517:39-54. [PMID: 39284539 DOI: 10.1016/j.ydbio.2024.09.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Revised: 08/16/2024] [Accepted: 09/08/2024] [Indexed: 09/29/2024]
Abstract
Combined removal of Crb1 and Crb2 from the developing optic vesicle evokes cellular and laminar disorganization by disrupting the apical cell-cell adhesion in developing retinal epithelium. As a result, at postnatal stages, affected mouse retinas show temporarily thickened, coarsely laminated retinas in addition to functional deficits, including a severely abnormal electroretinogram and decreased visual acuity. These features are reminiscent of Leber congenital amaurosis 8, which is caused in humans by subsets of Crb1 mutations. However, the cellular basis of the abnormalities in retinal progenitor cells (RPCs) that lead to retinal disorganization is largely unknown. In this study, we analyze specific features of RPCs in mutant retinas, including maintenance of the progenitor pool, cell cycle progression, cell cycle phase-dependent nuclear positioning, cell survival, and generation of mature retinal cell types. We find crucial defects in the mutant RPCs. Upon removal of CRB1 and CRB2, apical structures of the RPCs, determined by markers of cilia and centrosomes, are basally shifted. In addition, the positioning of the somata of the M-phase cells, normally localized at the apical surface of the retinal epithelium, is basally shifted in a nearly randomized pattern along the apico-basal axis. Consequently, we propose that positioning of RPCs is desynchronized from cell cycle phase and largely randomized during embryonic development at E17.5. Because the resultant postmitotic cells inevitably lose positional information, the outer and inner nuclear layers (ONL and INL) fail to form from ONBL during neonatal development and retinal cells become mixed locally and globally. Additional results of the lost tissue polarity in Crb1/Crb2 dKO retinas include atypical formation of heterotopic cell patches containing photoreceptor cells in the ganglion cell layer and acellular patches filled with neural processes. Collectively, these changes lead to a mouse model of LCA8-like pathology. LCA8-like pathology differs substantially from the well-characterized, broad range of degeneration phenotypes that arise during the differentiation of photoreceptor and Muller glial cells in retinitis pigmentosa 12, a closely related disease caused by mutated human Crb1. Importantly, the present results suggest that Crb1/Crb2 serve indispensable functions in maintaining cell-cycle phase-dependent positioning of RPCs along the apico-basal axis, regulating cell cycle progression, and maintaining structural laminar integrity without significantly affecting the size of the RPC pools, generation of the subsets of the retinal cell types, or the distribution of cell cycle phases during RPC division. Taken together, these findings provide the crucial cellular basis of the thickening and severely disorganized lamination that are the unique features of the retinal abnormalities in LCA8 patients.
Collapse
Affiliation(s)
- Seo-Hee Cho
- Center for Translational Medicine, Department of Medicine, Sydney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, 19107, USA.
| | - Ji Hyang Kim
- Center for Translational Medicine, Department of Medicine, Sydney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, 19107, USA
| | - Seonhee Kim
- Department of Neural Sciences, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, 19140, USA
| |
Collapse
|
3
|
Falnikar A, Quintremil S, Zhao HJ, Cheng HY, Helmer P, Tsai JW, Vallee RB. The nucleoporin Nup153 is the anchor for Kif1a during basal nuclear migration in brain progenitor cells. Cell Rep 2024; 43:115008. [PMID: 39666457 PMCID: PMC11702353 DOI: 10.1016/j.celrep.2024.115008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Revised: 10/14/2024] [Accepted: 11/07/2024] [Indexed: 12/14/2024] Open
Abstract
Radial glial progenitors (RGPs) are highly elongated epithelial cells that give rise to most stem cells, neurons, and glia in the vertebrate cerebral cortex. During development, the RGP nuclei exhibit a striking pattern of cell-cycle-dependent oscillatory movements known as interkinetic nuclear migration (INM), which we previously found to be mediated during G1 by the kinesin Kif1a and during G2 by cytoplasmic dynein, recruited to the nuclear envelope by the nucleoporins RanBP2 and Nup133. We now identify Nup153 as a nucleoporin anchor for Kif1a, responsible for G1-specific basal nuclear migration, providing a complete model for the mechanisms underlying this basic but mysterious behavior, with broad implications for understanding brain development.
Collapse
Affiliation(s)
- Aditi Falnikar
- Department of Pathology and Cell Biology, Columbia University Medical Center, New York, NY 10032, USA.
| | - Sebastian Quintremil
- Department of Pathology and Cell Biology, Columbia University Medical Center, New York, NY 10032, USA
| | - Hung-Jun Zhao
- Institute of Brain Science, College of Medicine, National Yang Ming Chiao Tung University, Taipei 112, Taiwan
| | - Haw-Yuan Cheng
- Institute of Brain Science, College of Medicine, National Yang Ming Chiao Tung University, Taipei 112, Taiwan
| | - Paige Helmer
- Department of Pathology and Cell Biology, Columbia University Medical Center, New York, NY 10032, USA
| | - Jin-Wu Tsai
- Institute of Brain Science, College of Medicine, National Yang Ming Chiao Tung University, Taipei 112, Taiwan
| | - Richard B Vallee
- Department of Pathology and Cell Biology, Columbia University Medical Center, New York, NY 10032, USA.
| |
Collapse
|
4
|
Yeung TJ, Wilkinson DG. Short-range Fgf signalling patterns hindbrain progenitors to induce the neurogenesis-to-oligodendrogenesis switch. Development 2024; 151:dev204256. [PMID: 39575980 DOI: 10.1242/dev.204256] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Accepted: 11/14/2024] [Indexed: 12/14/2024]
Abstract
In the vertebrate nervous system, neurogenesis generally precedes gliogenesis. The mechanisms driving the switch in cell type production and generation of the correct proportion of cell types remain unclear. Here, we show that Fgf20 signalling patterns progenitors to induce the switch from neurogenesis to oligodendrogenesis in the zebrafish hindbrain. Fgf20 emanating from earlier-born neurons signals at a short range to downregulate proneural gene expression in the segment centre with high spatial precision along both anterior-posterior and dorsal-ventral axes. This signal induces oligodendrocytes in the segment centre by upregulating olig2 and sox10 expression in pre-patterned competent progenitors. We show that the magnitude of proneural gene downregulation and the quantity of oligodendrocyte precursor cells specified is dependent on the extent of Fgf20 signalling. Overexpression of fgf20a induces precocious specification and differentiation of oligodendrocytes among olig2+ progenitors, resulting in an increase in oligodendrocytes at the expense of neurogenesis. Thus, Fgf20 signalling defines the proportion of each cell type produced. Taken together, Fgf20 signalling from earlier-born neurons patterns hindbrain segments spatially and temporally to induce the neurogenesis-to-oligodendrogenesis switch.
Collapse
Affiliation(s)
- Tim J Yeung
- The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | | |
Collapse
|
5
|
de Leeuw NF, Budhathoki R, Russell LJ, Loerke D, Blankenship JT. Nuclei as mechanical bumpers during epithelial remodeling. J Cell Biol 2024; 223:e202405078. [PMID: 39325019 PMCID: PMC11450824 DOI: 10.1083/jcb.202405078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Revised: 08/21/2024] [Accepted: 09/03/2024] [Indexed: 09/27/2024] Open
Abstract
The morphogenesis of developing tissues relies on extensive cellular rearrangements in shape, position, and identity. A key process in reshaping tissues is cell intercalation-driven elongation, where epithelial cells align and intercalate along a common axis. Typically, analyses focus on how peripheral cortical forces influence cell shape changes. Less attention is given to how inhomogeneities in internal structures, particularly the nucleus, impact cell shaping. Here, we examine how pulsed contractile and extension dynamics interact with the nucleus in elongating Drosophila embryos. Our data show that tightly packed nuclei in apical layers hinder tissue remodeling/oscillatory behaviors. We identify two mechanisms for resolving internuclear tensions: nuclear deformation and dispersion. Embryos with non-deformable nuclei use nuclear dispersion to maintain near-normal extensile rates, while those with non-dispersible nuclei due to microtubule inhibition exhibit disruptions in contractile behaviors. Disrupting both mechanisms leads to severe tissue extension defects and cell extrusion. These findings highlight the critical role of nuclear shape and positioning in topological remodeling of epithelia.
Collapse
Affiliation(s)
- Noah F. de Leeuw
- Department of Physics and Astronomy, University of Denver, Denver, CO, USA
| | - Rashmi Budhathoki
- Department of Biological Sciences, University of Denver, Denver, CO, USA
| | - Liam J. Russell
- Department of Physics and Astronomy, University of Denver, Denver, CO, USA
- Department of Biological Sciences, University of Denver, Denver, CO, USA
| | - Dinah Loerke
- Department of Physics and Astronomy, University of Denver, Denver, CO, USA
| | | |
Collapse
|
6
|
Wang S, He Q, Qu Y, Yin W, Zhao R, Wang X, Yang Y, Guo ZN. Emerging strategies for nerve repair and regeneration in ischemic stroke: neural stem cell therapy. Neural Regen Res 2024; 19:2430-2443. [PMID: 38526280 PMCID: PMC11090435 DOI: 10.4103/1673-5374.391313] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Revised: 09/26/2023] [Accepted: 11/10/2023] [Indexed: 03/26/2024] Open
Abstract
Ischemic stroke is a major cause of mortality and disability worldwide, with limited treatment options available in clinical practice. The emergence of stem cell therapy has provided new hope to the field of stroke treatment via the restoration of brain neuron function. Exogenous neural stem cells are beneficial not only in cell replacement but also through the bystander effect. Neural stem cells regulate multiple physiological responses, including nerve repair, endogenous regeneration, immune function, and blood-brain barrier permeability, through the secretion of bioactive substances, including extracellular vesicles/exosomes. However, due to the complex microenvironment of ischemic cerebrovascular events and the low survival rate of neural stem cells following transplantation, limitations in the treatment effect remain unresolved. In this paper, we provide a detailed summary of the potential mechanisms of neural stem cell therapy for the treatment of ischemic stroke, review current neural stem cell therapeutic strategies and clinical trial results, and summarize the latest advancements in neural stem cell engineering to improve the survival rate of neural stem cells. We hope that this review could help provide insight into the therapeutic potential of neural stem cells and guide future scientific endeavors on neural stem cells.
Collapse
Affiliation(s)
- Siji Wang
- Stroke Center, Department of Neurology, the First Hospital of Jilin University, Changchun, Jilin Province, China
| | - Qianyan He
- Stroke Center, Department of Neurology, the First Hospital of Jilin University, Changchun, Jilin Province, China
| | - Yang Qu
- Stroke Center, Department of Neurology, the First Hospital of Jilin University, Changchun, Jilin Province, China
| | - Wenjing Yin
- Stroke Center, Department of Neurology, the First Hospital of Jilin University, Changchun, Jilin Province, China
| | - Ruoyu Zhao
- Stroke Center, Department of Neurology, the First Hospital of Jilin University, Changchun, Jilin Province, China
| | - Xuyutian Wang
- Department of Breast Surgery, General Surgery Center, the First Hospital of Jilin University, Changchun, Jilin Province, China
| | - Yi Yang
- Stroke Center, Department of Neurology, the First Hospital of Jilin University, Changchun, Jilin Province, China
- Neuroscience Research Center, Department of Neurology, the First Hospital of Jilin University, Changchun, Jilin Province, China
| | - Zhen-Ni Guo
- Stroke Center, Department of Neurology, the First Hospital of Jilin University, Changchun, Jilin Province, China
- Neuroscience Research Center, Department of Neurology, the First Hospital of Jilin University, Changchun, Jilin Province, China
| |
Collapse
|
7
|
Willnow P, Teleman AA. Nuclear position and local acetyl-CoA production regulate chromatin state. Nature 2024; 630:466-474. [PMID: 38839952 PMCID: PMC11168921 DOI: 10.1038/s41586-024-07471-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Accepted: 04/25/2024] [Indexed: 06/07/2024]
Abstract
Histone acetylation regulates gene expression, cell function and cell fate1. Here we study the pattern of histone acetylation in the epithelial tissue of the Drosophila wing disc. H3K18ac, H4K8ac and total lysine acetylation are increased in the outer rim of the disc. This acetylation pattern is controlled by nuclear position, whereby nuclei continuously move from apical to basal locations within the epithelium and exhibit high levels of H3K18ac when they are in proximity to the tissue surface. These surface nuclei have increased levels of acetyl-CoA synthase, which generates the acetyl-CoA for histone acetylation. The carbon source for histone acetylation in the rim is fatty acid β-oxidation, which is also increased in the rim. Inhibition of fatty acid β-oxidation causes H3K18ac levels to decrease in the genomic proximity of genes involved in disc development. In summary, there is a physical mark of the outer rim of the wing and other imaginal epithelia in Drosophila that affects gene expression.
Collapse
Affiliation(s)
- Philipp Willnow
- German Cancer Research Center (DKFZ), Heidelberg, Germany
- Heidelberg University, Heidelberg, Germany
| | - Aurelio A Teleman
- German Cancer Research Center (DKFZ), Heidelberg, Germany.
- Heidelberg University, Heidelberg, Germany.
| |
Collapse
|
8
|
Tsai MH, Ke HC, Lin WC, Nian FS, Huang CW, Cheng HY, Hsu CS, Granata T, Chang CH, Castellotti B, Lin SY, Doniselli FM, Lu CJ, Franceschetti S, Ragona F, Hou PS, Canafoglia L, Tung CY, Lee MH, Wang WJ, Tsai JW. Novel lissencephaly-associated NDEL1 variant reveals distinct roles of NDE1 and NDEL1 in nucleokinesis and human cortical malformations. Acta Neuropathol 2024; 147:13. [PMID: 38194050 PMCID: PMC10776482 DOI: 10.1007/s00401-023-02665-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Revised: 11/23/2023] [Accepted: 11/24/2023] [Indexed: 01/10/2024]
Abstract
The development of the cerebral cortex involves a series of dynamic events, including cell proliferation and migration, which rely on the motor protein dynein and its regulators NDE1 and NDEL1. While the loss of function in NDE1 leads to microcephaly-related malformations of cortical development (MCDs), NDEL1 variants have not been detected in MCD patients. Here, we identified two patients with pachygyria, with or without subcortical band heterotopia (SBH), carrying the same de novo somatic mosaic NDEL1 variant, p.Arg105Pro (p.R105P). Through single-cell RNA sequencing and spatial transcriptomic analysis, we observed complementary expression of Nde1/NDE1 and Ndel1/NDEL1 in neural progenitors and post-mitotic neurons, respectively. Ndel1 knockdown by in utero electroporation resulted in impaired neuronal migration, a phenotype that could not be rescued by p.R105P. Remarkably, p.R105P expression alone strongly disrupted neuronal migration, increased the length of the leading process, and impaired nucleus-centrosome coupling, suggesting a failure in nucleokinesis. Mechanistically, p.R105P disrupted NDEL1 binding to the dynein regulator LIS1. This study identifies the first lissencephaly-associated NDEL1 variant and sheds light on the distinct roles of NDE1 and NDEL1 in nucleokinesis and MCD pathogenesis.
Collapse
Affiliation(s)
- Meng-Han Tsai
- Department of Neurology, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan
- School of Medicine, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Hao-Chen Ke
- Institute of Brain Science, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Department of Medical Education, Linkou Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Wan-Cian Lin
- Institute of Brain Science, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Faculty of Medicine, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Fang-Shin Nian
- Institute of Brain Science, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Institute of Clinical Medicine, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Chia-Wei Huang
- Institute of Brain Science, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Institute of Clinical Medicine, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Advanced Therapeutics Research Center, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Haw-Yuan Cheng
- Institute of Brain Science, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Chi-Sin Hsu
- Genomics Center for Clinical and Biotechnological Applications, Cancer Progression Research Center, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Tiziana Granata
- Department of Paediatric Neuroscience, European Reference Network EPIcare, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Chien-Hui Chang
- Institute of Brain Science, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Barbara Castellotti
- Unit of Medical Genetics and Neurogenetics, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Shin-Yi Lin
- Department of Biotechnology and Laboratory Science in Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Fabio M Doniselli
- Neuroradiology Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Cheng-Ju Lu
- Faculty of Medicine, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Silvana Franceschetti
- Integrated Diagnostics for Epilepsy, Department of Diagnostic and Technology, European Reference Network EPIcare, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Francesca Ragona
- Department of Paediatric Neuroscience, European Reference Network EPIcare, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Pei-Shan Hou
- Institute of Anatomy and Cell Biology, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Brain Research Center, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Laura Canafoglia
- Integrated Diagnostics for Epilepsy, Department of Diagnostic and Technology, European Reference Network EPIcare, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Chien-Yi Tung
- Advanced Therapeutics Research Center, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Mei-Hsuan Lee
- Institute of Clinical Medicine, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Advanced Therapeutics Research Center, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Won-Jing Wang
- Advanced Therapeutics Research Center, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Institute of Biochemistry and Molecule Biology, College of Life Science, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Jin-Wu Tsai
- Institute of Brain Science, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan.
- Advanced Therapeutics Research Center, National Yang Ming Chiao Tung University, Taipei, Taiwan.
- Brain Research Center, National Yang Ming Chiao Tung University, Taipei, Taiwan.
- Department of Biological Science and Technology, College of Biological Science and Technology, National Yang Ming Chiao Tung University, Hsinchu, Taiwan.
| |
Collapse
|
9
|
Pio T, Hill EJ, Kebede N, Andersen J, Sloan SA. Neuron-Astrocyte Interactions: A Human Perspective. ADVANCES IN NEUROBIOLOGY 2024; 39:69-93. [PMID: 39190072 DOI: 10.1007/978-3-031-64839-7_4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/28/2024]
Abstract
This chapter explores the intricate interactions between neurons and astrocytes within the nervous system with a particular emphasis on studies conducted in human tissue or with human cells. We specifically explore how neuron-astrocyte interactions relate to processes of cellular development, morphology, migration, synapse formation, and metabolism. These findings enrich our understanding of basic neurobiology and how disruptions in these processes are relevant to human diseases.The study of human neuron-astrocyte interactions is made possible because of transformative in vitro advancements that have facilitated the generation and sustained culture of human neural cells. In addition, the rise of techniques like sequencing at single-cell resolution has enabled the exploration of numerous human cell atlases and their comparisons to other animal model systems. Thus, the innovations outlined in this chapter illuminate the convergence and divergence of neuron-astrocyte interactions across species. As technologies progress, continually more sophisticated in vitro systems will increasingly reflect in vivo environments and deepen our command of neuron-glial interactions in human biology.
Collapse
Affiliation(s)
- Taylor Pio
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA, USA
| | - Emily J Hill
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA, USA
| | - Nardos Kebede
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA, USA
| | - Jimena Andersen
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA, USA
| | - Steven A Sloan
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA, USA.
| |
Collapse
|
10
|
Helmer P, Vallee RB. A two-kinesin mechanism controls neurogenesis in the developing brain. Commun Biol 2023; 6:1219. [PMID: 38040957 PMCID: PMC10692124 DOI: 10.1038/s42003-023-05604-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Accepted: 11/17/2023] [Indexed: 12/03/2023] Open
Abstract
During the course of brain development, Radial Glial Progenitor (RGP) cells give rise to most of the neurons required for a functional cortex. RGPs can undergo symmetric divisions, which result in RGP duplication, or asymmetric divisions, which result in one RGP as well as one to four neurons. The control of this balance is not fully understood, but must be closely regulated to produce the cells required for a functioning cortex, and to maintain the stem cell pool. In this study, we show that the balance between symmetric and asymmetric RGP divisions is in part regulated by the actions of two kinesins, Kif1A and Kif13B, which we find have opposing roles in neurogenesis through their action on the mitotic spindle in dividing RGPs. We find that Kif1A promotes neurogenesis, whereas Kif13B promotes symmetric, non-neurogenic divisions. Interestingly, the two kinesins are closely related in structure, and members of the same kinesin-3 subfamily, thus their opposing effects on spindle orientation appear to represent a novel mechanism for the regulation of neurogenesis.
Collapse
Affiliation(s)
- Paige Helmer
- Department of Pathology and Cell Biology, Columbia University Medical Center, New York, NY, 10032, USA.
- Department of Biological Sciences, Columbia University, New York, NY, 10032, USA.
| | - Richard B Vallee
- Department of Pathology and Cell Biology, Columbia University Medical Center, New York, NY, 10032, USA.
- Department of Biological Sciences, Columbia University, New York, NY, 10032, USA.
| |
Collapse
|
11
|
Han JS, Fishman-Williams E, Decker SC, Hino K, Reyes RV, Brown NL, Simó S, Torre AL. Notch directs telencephalic development and controls neocortical neuron fate determination by regulating microRNA levels. Development 2023; 150:dev201408. [PMID: 37272771 PMCID: PMC10309580 DOI: 10.1242/dev.201408] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Accepted: 04/28/2023] [Indexed: 05/13/2023]
Abstract
The central nervous system contains a myriad of different cell types produced from multipotent neural progenitors. Neural progenitors acquire distinct cell identities depending on their spatial position, but they are also influenced by temporal cues to give rise to different cell populations over time. For instance, the progenitors of the cerebral neocortex generate different populations of excitatory projection neurons following a well-known sequence. The Notch signaling pathway plays crucial roles during this process, but the molecular mechanisms by which Notch impacts progenitor fate decisions have not been fully resolved. Here, we show that Notch signaling is essential for neocortical and hippocampal morphogenesis, and for the development of the corpus callosum and choroid plexus. Our data also indicate that, in the neocortex, Notch controls projection neuron fate determination through the regulation of two microRNA clusters that include let-7, miR-99a/100 and miR-125b. Our findings collectively suggest that balanced Notch signaling is crucial for telencephalic development and that the interplay between Notch and miRNAs is essential for the control of neocortical progenitor behaviors and neuron cell fate decisions.
Collapse
Affiliation(s)
- Jisoo S. Han
- Department of Cell Biology and Human Anatomy, University of California Davis, Davis, CA 95616, USA
| | | | - Steven C. Decker
- Department of Cell Biology and Human Anatomy, University of California Davis, Davis, CA 95616, USA
| | - Keiko Hino
- Department of Cell Biology and Human Anatomy, University of California Davis, Davis, CA 95616, USA
| | - Raenier V. Reyes
- Department of Cell Biology and Human Anatomy, University of California Davis, Davis, CA 95616, USA
| | - Nadean L. Brown
- Department of Cell Biology and Human Anatomy, University of California Davis, Davis, CA 95616, USA
| | - Sergi Simó
- Department of Cell Biology and Human Anatomy, University of California Davis, Davis, CA 95616, USA
| | - Anna La Torre
- Department of Cell Biology and Human Anatomy, University of California Davis, Davis, CA 95616, USA
| |
Collapse
|
12
|
Yoshida MW, Hakozaki M, Goshima G. Armadillo repeat-containing kinesin represents the versatile plus-end-directed transporter in Physcomitrella. NATURE PLANTS 2023; 9:733-748. [PMID: 37142749 DOI: 10.1038/s41477-023-01397-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Accepted: 03/21/2023] [Indexed: 05/06/2023]
Abstract
Kinesin-1, also known as conventional kinesin, is widely used for microtubule plus-end-directed (anterograde) transport of various cargos in animal cells. However, a motor functionally equivalent to the conventional kinesin has not been identified in plants, which lack the kinesin-1 genes. Here we show that plant-specific armadillo repeat-containing kinesin (ARK) is the long sought-after versatile anterograde transporter in plants. In ARK mutants of the moss Physcomitrium patens, the anterograde motility of nuclei, chloroplasts, mitochondria and secretory vesicles was suppressed. Ectopic expression of non-motile or tail-deleted ARK did not restore organelle distribution. Another prominent macroscopic phenotype of ARK mutants was the suppression of cell tip growth. We showed that this defect was attributed to the mislocalization of actin regulators, including RopGEFs; expression and forced apical localization of RopGEF3 partially rescued the growth phenotype of the ARK mutant. The mutant phenotypes were partially rescued by ARK homologues in Arabidopsis thaliana, suggesting the conservation of ARK functions in plants.
Collapse
Affiliation(s)
- Mari W Yoshida
- Department of Biological Science, Graduate School of Science, Nagoya University, Nagoya, Japan
| | - Maya Hakozaki
- Department of Biological Science, Graduate School of Science, Nagoya University, Nagoya, Japan
| | - Gohta Goshima
- Department of Biological Science, Graduate School of Science, Nagoya University, Nagoya, Japan.
- Sugashima Marine Biological Laboratory, Graduate School of Science, Nagoya University, Toba, Japan.
| |
Collapse
|
13
|
Napoli FR, Daly CM, Neal S, McCulloch KJ, Zaloga AR, Liu A, Koenig KM. Cephalopod retinal development shows vertebrate-like mechanisms of neurogenesis. Curr Biol 2022; 32:5045-5056.e3. [PMID: 36356573 PMCID: PMC9729453 DOI: 10.1016/j.cub.2022.10.027] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Revised: 09/30/2022] [Accepted: 10/14/2022] [Indexed: 11/10/2022]
Abstract
Coleoid cephalopods, including squid, cuttlefish, and octopus, have large and complex nervous systems and high-acuity, camera-type eyes. These traits are comparable only to features that are independently evolved in the vertebrate lineage. The size of animal nervous systems and the diversity of their constituent cell types is a result of the tight regulation of cellular proliferation and differentiation in development. Changes in the process of development during evolution that result in a diversity of neural cell types and variable nervous system size are not well understood. Here, we have pioneered live-imaging techniques and performed functional interrogation to show that the squid Doryteuthis pealeii utilizes mechanisms during retinal neurogenesis that are hallmarks of vertebrate processes. We find that retinal progenitor cells in the squid undergo nuclear migration until they exit the cell cycle. We identify retinal organization corresponding to progenitor, post-mitotic, and differentiated cells. Finally, we find that Notch signaling may regulate both retinal cell cycle and cell fate. Given the convergent evolution of elaborate visual systems in cephalopods and vertebrates, these results reveal common mechanisms that underlie the growth of highly proliferative neurogenic primordia. This work highlights mechanisms that may alter ontogenetic allometry and contribute to the evolution of complexity and growth in animal nervous systems.
Collapse
Affiliation(s)
- Francesca R Napoli
- John Harvard Distinguished Science Fellowship Program, Harvard University, Cambridge, MA 02138, USA; Department of Organismic and Evolutionary Biology, Harvard University, Harvard University, Cambridge, MA 02138, USA
| | - Christina M Daly
- John Harvard Distinguished Science Fellowship Program, Harvard University, Cambridge, MA 02138, USA; Department of Organismic and Evolutionary Biology, Harvard University, Harvard University, Cambridge, MA 02138, USA
| | - Stephanie Neal
- John Harvard Distinguished Science Fellowship Program, Harvard University, Cambridge, MA 02138, USA; Department of Organismic and Evolutionary Biology, Harvard University, Harvard University, Cambridge, MA 02138, USA
| | - Kyle J McCulloch
- John Harvard Distinguished Science Fellowship Program, Harvard University, Cambridge, MA 02138, USA; Department of Organismic and Evolutionary Biology, Harvard University, Harvard University, Cambridge, MA 02138, USA
| | - Alexandra R Zaloga
- John Harvard Distinguished Science Fellowship Program, Harvard University, Cambridge, MA 02138, USA; Department of Organismic and Evolutionary Biology, Harvard University, Harvard University, Cambridge, MA 02138, USA
| | - Alicia Liu
- John Harvard Distinguished Science Fellowship Program, Harvard University, Cambridge, MA 02138, USA; Department of Organismic and Evolutionary Biology, Harvard University, Harvard University, Cambridge, MA 02138, USA
| | - Kristen M Koenig
- John Harvard Distinguished Science Fellowship Program, Harvard University, Cambridge, MA 02138, USA; Department of Organismic and Evolutionary Biology, Harvard University, Harvard University, Cambridge, MA 02138, USA.
| |
Collapse
|
14
|
Fischer NC, Friedman V, Martinez-Reyes MA, Hao H, Chowdhury TA, Starr DA, Quinn CC. The ANC-1 (Nesprin-1/2) organelle-anchoring protein functions through mitochondria to polarize axon growth in response to SLT-1. PLoS Genet 2022; 18:e1010521. [PMID: 36409768 PMCID: PMC9721489 DOI: 10.1371/journal.pgen.1010521] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Revised: 12/05/2022] [Accepted: 11/11/2022] [Indexed: 11/22/2022] Open
Abstract
A family of giant KASH proteins, including C. elegans ANC-1 and mammalian Nesprin-1 and -2, are involved in organelle anchoring and are associated with multiple neurodevelopmental disorders including autism, bipolar disorder, and schizophrenia. However, little is known about how these proteins function in neurons. Moreover, the role of organelle anchoring in axon development is poorly understood. Here, we report that ANC-1 functions with the SLT-1 extracellular guidance cue to polarize ALM axon growth. This role for ANC-1 is specific to its longer ANC-1A and ANC-1C isoforms, suggesting that it is mechanistically distinct from previously described roles for ANC-1. We find that ANC-1 is required for the localization of a cluster of mitochondria to the base of the proximal axon. Furthermore, genetic and pharmacological studies indicate that ANC-1 functions with mitochondria to promote polarization of ALM axon growth. These observations reveal a mechanism whereby ANC-1 functions through mitochondria to polarize axon growth in response to SLT-1.
Collapse
Affiliation(s)
- Nathan C. Fischer
- Department of Biological Sciences, University of Wisconsin-Milwaukee; Milwaukee, Wisconsin, United States of America
| | - Vladislav Friedman
- Department of Biological Sciences, University of Wisconsin-Milwaukee; Milwaukee, Wisconsin, United States of America
| | - Miguel A. Martinez-Reyes
- Department of Biological Sciences, University of Wisconsin-Milwaukee; Milwaukee, Wisconsin, United States of America
| | - Hongyan Hao
- Department of Molecular and Cellular Biology, University of California, Davis, California, United States of America
| | - Tamjid A. Chowdhury
- Department of Biological Sciences, University of Wisconsin-Milwaukee; Milwaukee, Wisconsin, United States of America
| | - Daniel A. Starr
- Department of Molecular and Cellular Biology, University of California, Davis, California, United States of America
| | - Christopher C. Quinn
- Department of Biological Sciences, University of Wisconsin-Milwaukee; Milwaukee, Wisconsin, United States of America
| |
Collapse
|
15
|
Kawano D, Pinter K, Chlebowski M, Petralia RS, Wang YX, Nechiporuk AV, Drerup CM. NudC regulated Lis1 stability is essential for the maintenance of dynamic microtubule ends in axon terminals. iScience 2022; 25:105072. [PMID: 36147950 PMCID: PMC9485903 DOI: 10.1016/j.isci.2022.105072] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Revised: 07/26/2022] [Accepted: 08/30/2022] [Indexed: 11/28/2022] Open
Abstract
In the axon terminal, microtubule stability is decreased relative to the axon shaft. The dynamic microtubule plus ends found in the axon terminal have many functions, including serving as a docking site for the Cytoplasmic dynein motor. Here, we report an unexplored function of dynein in microtubule regulation in axon terminals: regulation of microtubule stability. Using a forward genetic screen, we identified a mutant with an abnormal axon terminal structure owing to a loss of function mutation in NudC. We show that, in the axon terminal, NudC is a chaperone for the protein Lis1. Decreased Lis1 in nudc axon terminals causes dynein/dynactin accumulation and increased microtubule stability. Microtubule dynamics can be restored by pharmacologically inhibiting dynein, implicating excess dynein motor function in microtubule stabilization. Together, our data support a model in which local NudC-Lis1 modulation of the dynein motor is critical for the regulation of microtubule stability in the axon terminal. NudC, a dynein regulator, is crucial for axon terminal structure NudC mutation leads to a near complete loss of Lis1 protein in axon terminals Lis1 deficits cause accumulation of dynein and cargo in axon terminals Local elevation of dynein increases axon terminal microtubule stability
Collapse
Affiliation(s)
- Dane Kawano
- Unit on Neuronal Cell Biology, National Institute of Child Health & Human Development, National Institutes of Health, Bethesda, MD 20892, USA
| | - Katherine Pinter
- Unit on Neuronal Cell Biology, National Institute of Child Health & Human Development, National Institutes of Health, Bethesda, MD 20892, USA
| | - Madison Chlebowski
- Department of Integrative Biology, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Ronald S Petralia
- Advanced Imaging Core, National Institute of Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, MD 20892, USA
| | - Ya-Xian Wang
- Advanced Imaging Core, National Institute of Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, MD 20892, USA
| | - Alex V Nechiporuk
- Department of Cell, Developmental & Cancer Biology, Oregon Health & Science University, Portland, OR 97239, USA
| | - Catherine M Drerup
- Unit on Neuronal Cell Biology, National Institute of Child Health & Human Development, National Institutes of Health, Bethesda, MD 20892, USA.,Department of Integrative Biology, University of Wisconsin-Madison, Madison, WI 53706, USA
| |
Collapse
|
16
|
Wang S, Fu Y, Miyata T, Matsumoto S, Shinoda T, Itoh K, Harada A, Hirotsune S, Jin M. Functional Cooperation of α-Synuclein and Tau Is Essential for Proper Corticogenesis. J Neurosci 2022; 42:7031-7046. [PMID: 35906071 PMCID: PMC9480882 DOI: 10.1523/jneurosci.0396-22.2022] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2022] [Revised: 07/07/2022] [Accepted: 07/21/2022] [Indexed: 11/21/2022] Open
Abstract
Alpha-synuclein (αSyn) and tau are abundant multifunctional neuronal proteins, and their intracellular deposits have been linked to many neurodegenerative diseases, including Alzheimer's disease and Parkinson's disease. Despite the disease relevance, their physiological roles remain elusive, as mice with knock-out of either of these genes do not exhibit overt phenotypes. To reveal functional cooperation, we generated αSyn-/-tau-/- double-knock-out mice and characterized the functional cross talk between these proteins during brain development. Intriguingly, deletion of αSyn and tau reduced Notch signaling and accelerated interkinetic nuclear migration of G2 phase at early embryonic stage. This significantly altered the balance between the proliferative and neurogenic divisions of progenitor cells, resulting in an overproduction of early born neurons and enhanced neurogenesis, by which the brain size was enlarged during the embryonic stage in both sexes. On the other hand, a reduction in the number of neural progenitor cells in the middle stage of corticogenesis diminished subsequent gliogenesis in the αSyn-/-tau-/- cortex. Additionally, the expansion and maturation of macroglial cells (astrocytes and oligodendrocytes) were suppressed in the αSyn-/-tau-/- postnatal brain, which in turn reduced the male αSyn-/-tau-/- brain size and cortical thickness to less than the control values. Our study identifies important functional cooperation of αSyn and tau during corticogenesis.SIGNIFICANCE STATEMENT Correct understanding of the physiological functions of αSyn and tau in CNS is critical to elucidate pathogenesis involved in the etiology of neurodegenerative diseases including Alzheimer's disease and Parkinson's disease. We show here that αSyn and tau are cooperatively involved in brain development via maintenance of progenitor cells. αSyn and tau double-knock-out mice exhibited an overproduction of early born neurons and accelerated neurogenesis at early corticogenesis. Furthermore, loss of αSyn and tau also perturbed gliogenesis at later embryonic stage, as well as the subsequent glial expansion and maturation at postnatal brain. Our findings provide new mechanistic insights and extend therapeutic opportunities for neurodegenerative diseases caused by aberrant αSyn and tau.
Collapse
Affiliation(s)
- Shengming Wang
- Department of Genetic Disease Research, Osaka Metropolitan University Graduate School of Medicine, Osaka 545-8585, Japan
| | - Yu Fu
- Department of Genetic Disease Research, Osaka Metropolitan University Graduate School of Medicine, Osaka 545-8585, Japan
| | - Takaki Miyata
- Department of Anatomy and Cell Biology, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
| | - Sakiko Matsumoto
- Department of Genetic Disease Research, Osaka Metropolitan University Graduate School of Medicine, Osaka 545-8585, Japan
| | - Tomoyasu Shinoda
- Department of Anatomy and Cell Biology, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
| | - Kyoko Itoh
- Department of Pathology and Applied Neurobiology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan
| | - Akihiro Harada
- Department of Cell Biology, Graduate School of Medicine, Osaka University, Osaka 565-0871, Japan
| | - Shinji Hirotsune
- Department of Genetic Disease Research, Osaka Metropolitan University Graduate School of Medicine, Osaka 545-8585, Japan
| | - Mingyue Jin
- Department of Genetic Disease Research, Osaka Metropolitan University Graduate School of Medicine, Osaka 545-8585, Japan
- Guangxi Key Laboratory of Brain and Cognitive Neuroscience, Guilin Medical University, Guilin, Guangxi 541199, China
| |
Collapse
|
17
|
Cheng L, Cring MR, Wadkins DA, Kuehn MH. Absence of Connexin 43 Results in Smaller Retinas and Arrested, Depolarized Retinal Progenitor Cells in Human Retinal Organoids. Stem Cells 2022; 40:592-604. [PMID: 35263762 DOI: 10.1093/stmcls/sxac017] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Accepted: 02/18/2022] [Indexed: 11/14/2022]
Abstract
The development of the vertebrate retina relies on complex regulatory mechanisms to achieve its characteristic layered morphology containing multiple neuronal cell types. While connexin 43 (CX43) is not expressed by mature retinal neurons, mutations in its gene GJA1 are associated with microphthalmia and low vision in patients. To delineate how lack of CX43 affects retinal development, GJA1 was disrupted in human induced pluripotent stem cells (hiPSCs) (GJA1-/-) using CRISPR/Cas9 editing, and these were subsequently differentiated into retinal organoids. GJA1-/- hiPSCs do not display defects in self-renewal and pluripotency, but the resulting organoids are smaller with a thinner neural retina and decreased abundance of many retinal cell types. CX43-deficient organoids express lower levels of the neural marker PAX6 and the retinal progenitor cell (RPC) markers PAX6, SIX3, and SIX6. Conversely, expression of the early neuroectoderm markers SOX1 and SOX2 remains high in GJA1-/- organoids throughout their development. The lack of CX43 results in an increased population of CHX10-positive RPCs that are smaller, disorganized, do not become polarized, and possess a limited ability to commit to retinal fate specification. Our data indicate that lack of CX43 causes a developmental arrest in RPCs that subsequently leads to pan-retinal defects and stunted ocular growth.
Collapse
Affiliation(s)
- Lin Cheng
- Department of Ophthalmology and Visual Sciences, University of Iowa Carver College of Medicine, Iowa City, IA, USA
- Center for the Prevention and Treatment of Visual Loss, Veterans Affairs Medical Center, Iowa City, IA, USA
| | - Matthew R Cring
- Department of Pediatrics, University of Iowa Carver College of Medicine, Iowa City, IA, USA
| | - David A Wadkins
- Department of Ophthalmology and Visual Sciences, University of Iowa Carver College of Medicine, Iowa City, IA, USA
- Center for the Prevention and Treatment of Visual Loss, Veterans Affairs Medical Center, Iowa City, IA, USA
| | - Markus H Kuehn
- Department of Ophthalmology and Visual Sciences, University of Iowa Carver College of Medicine, Iowa City, IA, USA
- Center for the Prevention and Treatment of Visual Loss, Veterans Affairs Medical Center, Iowa City, IA, USA
- Institute for Vision Research, University of Iowa Carver College of Medicine, Iowa City, IA, USA
| |
Collapse
|
18
|
A Nuclear Belt Fastens on Neural Cell Fate. Cells 2022; 11:cells11111761. [PMID: 35681456 PMCID: PMC9179901 DOI: 10.3390/cells11111761] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Revised: 05/20/2022] [Accepted: 05/21/2022] [Indexed: 12/22/2022] Open
Abstract
Successful embryonic and adult neurogenesis require proliferating neural stem and progenitor cells that are intrinsically and extrinsically guided into a neuronal fate. In turn, migration of new-born neurons underlies the complex cytoarchitecture of the brain. Proliferation and migration are therefore essential for brain development, homeostasis and function in adulthood. Among several tightly regulated processes involved in brain formation and function, recent evidence points to the nuclear envelope (NE) and NE-associated components as critical new contributors. Classically, the NE was thought to merely represent a barrier mediating selective exchange between the cytoplasm and nucleoplasm. However, research over the past two decades has highlighted more sophisticated and diverse roles for NE components in progenitor fate choice and migration of their progeny by tuning gene expression via interactions with chromatin, transcription factors and epigenetic factors. Defects in NE components lead to neurodevelopmental impairments, whereas age-related changes in NE components are proposed to influence neurodegenerative diseases. Thus, understanding the roles of NE components in brain development, maintenance and aging is likely to reveal new pathophysiological mechanisms for intervention. Here, we review recent findings for the previously underrepresented contribution of the NE in neuronal commitment and migration, and envision future avenues for investigation.
Collapse
|
19
|
Vaid S, Huttner WB. Progenitor-Based Cell Biological Aspects of Neocortex Development and Evolution. Front Cell Dev Biol 2022; 10:892922. [PMID: 35602606 PMCID: PMC9119302 DOI: 10.3389/fcell.2022.892922] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Accepted: 04/14/2022] [Indexed: 11/13/2022] Open
Abstract
During development, the decision of stem and progenitor cells to switch from proliferation to differentiation is of critical importance for the overall size of an organ. Too early a switch will deplete the stem/progenitor cell pool, and too late a switch will not generate the required differentiated cell types. With a focus on the developing neocortex, a six-layered structure constituting the major part of the cerebral cortex in mammals, we discuss here the cell biological features that are crucial to ensure the appropriate proliferation vs. differentiation decision in the neural progenitor cells. In the last two decades, the neural progenitor cells giving rise to the diverse types of neurons that function in the neocortex have been intensely investigated for their role in cortical expansion and gyrification. In this review, we will first describe these different progenitor types and their diversity. We will then review the various cell biological features associated with the cell fate decisions of these progenitor cells, with emphasis on the role of the radial processes emanating from these progenitor cells. We will also discuss the species-specific differences in these cell biological features that have allowed for the evolutionary expansion of the neocortex in humans. Finally, we will discuss the emerging role of cell cycle parameters in neocortical expansion.
Collapse
Affiliation(s)
- Samir Vaid
- Department of Basic Neurosciences, University of Geneva, Geneva, Switzerland
- *Correspondence: Samir Vaid, ; Wieland B. Huttner,
| | - Wieland B. Huttner
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
- *Correspondence: Samir Vaid, ; Wieland B. Huttner,
| |
Collapse
|
20
|
Romero-Morales AI, Gama V. Revealing the Impact of Mitochondrial Fitness During Early Neural Development Using Human Brain Organoids. Front Mol Neurosci 2022; 15:840265. [PMID: 35571368 PMCID: PMC9102998 DOI: 10.3389/fnmol.2022.840265] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Accepted: 04/04/2022] [Indexed: 11/13/2022] Open
Abstract
Mitochondrial homeostasis -including function, morphology, and inter-organelle communication- provides guidance to the intrinsic developmental programs of corticogenesis, while also being responsive to environmental and intercellular signals. Two- and three-dimensional platforms have become useful tools to interrogate the capacity of cells to generate neuronal and glia progeny in a background of metabolic dysregulation, but the mechanistic underpinnings underlying the role of mitochondria during human neurogenesis remain unexplored. Here we provide a concise overview of cortical development and the use of pluripotent stem cell models that have contributed to our understanding of mitochondrial and metabolic regulation of early human brain development. We finally discuss the effects of mitochondrial fitness dysregulation seen under stress conditions such as metabolic dysregulation, absence of developmental apoptosis, and hypoxia; and the avenues of research that can be explored with the use of brain organoids.
Collapse
Affiliation(s)
| | - Vivian Gama
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN, United States
- Vanderbilt Center for Stem Cell Biology, Vanderbilt University, Nashville, TN, United States
- Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN, United States
| |
Collapse
|
21
|
Garrott SR, Gillies JP, DeSantis ME. Nde1 and Ndel1: Outstanding Mysteries in Dynein-Mediated Transport. Front Cell Dev Biol 2022; 10:871935. [PMID: 35493069 PMCID: PMC9041303 DOI: 10.3389/fcell.2022.871935] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Accepted: 03/17/2022] [Indexed: 11/17/2022] Open
Abstract
Cytoplasmic dynein-1 (dynein) is the primary microtubule minus-end directed molecular motor in most eukaryotes. As such, dynein has a broad array of functions that range from driving retrograde-directed cargo trafficking to forming and focusing the mitotic spindle. Dynein does not function in isolation. Instead, a network of regulatory proteins mediate dynein’s interaction with cargo and modulate dynein’s ability to engage with and move on the microtubule track. A flurry of research over the past decade has revealed the function and mechanism of many of dynein’s regulators, including Lis1, dynactin, and a family of proteins called activating adaptors. However, the mechanistic details of two of dynein’s important binding partners, the paralogs Nde1 and Ndel1, have remained elusive. While genetic studies have firmly established Nde1/Ndel1 as players in the dynein transport pathway, the nature of how they regulate dynein activity is unknown. In this review, we will compare Ndel1 and Nde1 with a focus on discerning if the proteins are functionally redundant, outline the data that places Nde1/Ndel1 in the dynein transport pathway, and explore the literature supporting and opposing the predominant hypothesis about Nde1/Ndel1’s molecular effect on dynein activity.
Collapse
Affiliation(s)
- Sharon R. Garrott
- Department of Biological Chemistry, University of Michigan, Ann Arbor, MI, United States
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI, United States
| | - John P. Gillies
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI, United States
| | - Morgan E. DeSantis
- Department of Biological Chemistry, University of Michigan, Ann Arbor, MI, United States
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI, United States
- *Correspondence: Morgan E. DeSantis,
| |
Collapse
|
22
|
Wang X, Sarver AL, Han Q, Seiler CL, Xie C, Lu H, Forster CL, Tretyakova NY, Hallstrom TC. UHRF2 regulates cell cycle, epigenetics and gene expression to control the timing of retinal progenitor and ganglion cell differentiation. Development 2022; 149:274710. [PMID: 35285483 PMCID: PMC8984156 DOI: 10.1242/dev.195644] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2020] [Accepted: 01/28/2022] [Indexed: 11/20/2022]
Abstract
Ubiquitin-like, containing PHD and RING finger domains 2 (UHRF2) regulates cell cycle and binds 5-hydroxymethylcytosine (5hmC) to promote completion of DNA demethylation. Uhrf2-/- mice are without gross phenotypic defects; however, the cell cycle and epigenetic regulatory functions of Uhrf2 during retinal tissue development are unclear. Retinal progenitor cells (RPCs) produce all retinal neurons and Müller glia in a predictable sequence controlled by the complex interplay between extrinsic signaling, cell cycle, epigenetic changes and cell-specific transcription factor activation. In this study, we find that UHRF2 accumulates in RPCs, and its conditional deletion from mouse RPCs reduced 5hmC, altered gene expressions and disrupted retinal cell proliferation and differentiation. Retinal ganglion cells were overproduced in Uhrf2-deficient retinae at the expense of VSX2+ RPCs. Most other cell types were transiently delayed in differentiation. Expression of each member of the Tet3/Uhrf2/Tdg active demethylation pathway was reduced in Uhrf2-deficient retinae, consistent with locally reduced 5hmC in their gene bodies. This study highlights a novel role of UHRF2 in controlling the transition from RPCs to differentiated cell by regulating cell cycle, epigenetic and gene expression decisions.
Collapse
Affiliation(s)
- Xiaohong Wang
- Department of Pediatrics, Division of Blood and Marrow Transplantation, 420 Delaware Street S.E., University of Minnesota, Minneapolis, MN 55455, USA
| | - Aaron L Sarver
- Institute for Health Informatics, Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA
| | - Qiyuan Han
- Department of Medicinal Chemistry, University of Minnesota, Minneapolis, MN 55455, USA
| | - Christopher L Seiler
- Department of Medicinal Chemistry, University of Minnesota, Minneapolis, MN 55455, USA
| | - Chencheng Xie
- Department of Pediatrics, Division of Blood and Marrow Transplantation, 420 Delaware Street S.E., University of Minnesota, Minneapolis, MN 55455, USA
| | - Huarui Lu
- Department of Pediatrics, Division of Blood and Marrow Transplantation, 420 Delaware Street S.E., University of Minnesota, Minneapolis, MN 55455, USA
| | - Colleen L Forster
- BioNet, Academic Health Center, University of Minnesota, Minneapolis, MN 55455, USA
| | - Natalia Y Tretyakova
- Department of Medicinal Chemistry, University of Minnesota, Minneapolis, MN 55455, USA
| | - Timothy C Hallstrom
- Department of Pediatrics, Division of Blood and Marrow Transplantation, 420 Delaware Street S.E., University of Minnesota, Minneapolis, MN 55455, USA
| |
Collapse
|
23
|
Pfeuty B. Multistability and transitions between spatiotemporal patterns through versatile Notch-Hes signaling. J Theor Biol 2022; 539:111060. [DOI: 10.1016/j.jtbi.2022.111060] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Revised: 01/02/2022] [Accepted: 02/08/2022] [Indexed: 10/19/2022]
|
24
|
Zaqout S, Kaindl AM. Autosomal Recessive Primary Microcephaly: Not Just a Small Brain. Front Cell Dev Biol 2022; 9:784700. [PMID: 35111754 PMCID: PMC8802810 DOI: 10.3389/fcell.2021.784700] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Accepted: 12/01/2021] [Indexed: 02/06/2023] Open
Abstract
Microcephaly or reduced head circumference results from a multitude of abnormal developmental processes affecting brain growth and/or leading to brain atrophy. Autosomal recessive primary microcephaly (MCPH) is the prototype of isolated primary (congenital) microcephaly, affecting predominantly the cerebral cortex. For MCPH, an accelerating number of mutated genes emerge annually, and they are involved in crucial steps of neurogenesis. In this review article, we provide a deeper look into the microcephalic MCPH brain. We explore cytoarchitecture focusing on the cerebral cortex and discuss diverse processes occurring at the level of neural progenitors, early generated and mature neurons, and glial cells. We aim to thereby give an overview of current knowledge in MCPH phenotype and normal brain growth.
Collapse
Affiliation(s)
- Sami Zaqout
- Department of Basic Medical Sciences, College of Medicine, QU Health, Qatar University, Doha, Qatar
- Biomedical and Pharmaceutical Research Unit, QU Health, Qatar University, Doha, Qatar
| | - Angela M. Kaindl
- Institute of Cell and Neurobiology, Charité—Universitätsmedizin Berlin, Berlin, Germany
- Center for Chronically Sick Children (Sozialpädiatrisches Zentrum, SPZ), Charité—Universitätsmedizin Berlin, Berlin, Germany
- Department of Pediatric Neurology, Charité—Universitätsmedizin Berlin, Berlin, Germany
| |
Collapse
|
25
|
Selvaggio G, Cristellon S, Marchetti L. A Novel Hybrid Logic-ODE Modeling Approach to Overcome Knowledge Gaps. Front Mol Biosci 2022; 8:760077. [PMID: 34988115 PMCID: PMC8721169 DOI: 10.3389/fmolb.2021.760077] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Accepted: 11/09/2021] [Indexed: 11/13/2022] Open
Abstract
Mathematical modeling allows using different formalisms to describe, investigate, and understand biological processes. However, despite the advent of high-throughput experimental techniques, quantitative information is still a challenge when looking for data to calibrate model parameters. Furthermore, quantitative formalisms must cope with stiffness and tractability problems, more so if used to describe multicellular systems. On the other hand, qualitative models may lack the proper granularity to describe the underlying kinetic processes. We propose a hybrid modeling approach that integrates ordinary differential equations and logical formalism to describe distinct biological layers and their communication. We focused on a multicellular system as a case study by applying the hybrid formalism to the well-known Delta-Notch signaling pathway. We used a differential equation model to describe the intracellular pathways while the cell-cell interactions were defined by logic rules. The hybrid approach herein employed allows us to combine the pros of different modeling techniques by overcoming the lack of quantitative information with a qualitative description that discretizes activation and inhibition processes, thus avoiding complexity.
Collapse
Affiliation(s)
- Gianluca Selvaggio
- Piazza Manifattura, Fondazione The Microsoft Research-University of Trento Centre for Computational and Systems Biology (COSBI), Rovereto, Italy
| | - Serena Cristellon
- Piazza Manifattura, Fondazione The Microsoft Research-University of Trento Centre for Computational and Systems Biology (COSBI), Rovereto, Italy.,Department of Mathematics, University of Trento, Trento, Italy
| | - Luca Marchetti
- Piazza Manifattura, Fondazione The Microsoft Research-University of Trento Centre for Computational and Systems Biology (COSBI), Rovereto, Italy.,Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, Trento, Italy
| |
Collapse
|
26
|
Han X, Wei Y, Ba R, Sun L, Zhao C. PDK1 Regulates the Lengthening of G1 Phase to Balance RGC Proliferation and Differentiation during Cortical Neurogenesis. Cereb Cortex 2021; 32:3488-3500. [PMID: 34918060 DOI: 10.1093/cercor/bhab428] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Revised: 11/02/2021] [Accepted: 11/02/2021] [Indexed: 12/11/2022] Open
Abstract
During cortical development, the balance between progenitor self-renewal and neurogenesis is critical for determining the size/morphology of the cortex. A fundamental feature of the developing cortex is an increase in the length of G1 phase in RGCs over the course of neurogenesis, which is a key determinant of progenitor fate choice. How the G1 length is temporally regulated remains unclear. Here, Pdk1, a member of the AGC kinase family, was conditionally disrupted by crossing an Emx1-Cre mouse line with a Pdk1fl/fl line. The loss of Pdk1 led to a shorter cell cycle accompanied by increased RGC proliferation specifically at late rather than early/middle neurogenic stages, which was attributed to impaired lengthening of G1 phase. Coincidently, apical-to-basal interkinetic nuclear migration was accelerated in Pdk1 cKO cortices. Consequently, we detected an increased neuronal output at P0. We further showed the significant upregulation of the cell cycle regulator cyclin D1 and its activator Myc in the cKO cortices relative to those of control animals. Overall, we have identified a novel role for PDK1 in cortical neurogenesis. PDK1 functions as an upstream regulator of the Myc-cyclin D1 pathway to control the lengthening of G1 phase and the balance between RGC proliferation and differentiation.
Collapse
Affiliation(s)
- Xiaoning Han
- Key Laboratory of Developmental Genes and Human Diseases, Ministry of Education, School of Medicine, Southeast University, Nanjing 210009, China.,Institute of Biomedical Engineering and Health Science, Changzhou University, Changzhou 213164, China
| | - Yongjie Wei
- Key Laboratory of Developmental Genes and Human Diseases, Ministry of Education, School of Medicine, Southeast University, Nanjing 210009, China
| | - Ru Ba
- Key Laboratory of Developmental Genes and Human Diseases, Ministry of Education, School of Medicine, Southeast University, Nanjing 210009, China
| | - Lijuan Sun
- Key Laboratory of Developmental Genes and Human Diseases, Ministry of Education, School of Medicine, Southeast University, Nanjing 210009, China
| | - Chunjie Zhao
- Key Laboratory of Developmental Genes and Human Diseases, Ministry of Education, School of Medicine, Southeast University, Nanjing 210009, China
| |
Collapse
|
27
|
Collins MA, Coon LA, Thomas R, Mandigo TR, Wynn E, Folker ES. Ensconsin-dependent changes in microtubule organization and LINC complex-dependent changes in nucleus-nucleus interactions result in quantitatively distinct myonuclear positioning defects. Mol Biol Cell 2021; 32:ar27. [PMID: 34524872 PMCID: PMC8693964 DOI: 10.1091/mbc.e21-06-0324] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
Nuclear movement is a fundamental process of eukaryotic cell biology. Skeletal muscle presents an intriguing model to study nuclear movement because its development requires the precise positioning of multiple nuclei within a single cytoplasm. Furthermore, there is a high correlation between aberrant nuclear positioning and poor muscle function. Although many genes that regulate nuclear movement have been identified, the mechanisms by which these genes act are not known. Using Drosophila melanogaster muscle development as a model system and a combination of live-embryo microscopy and laser ablation of nuclei, we have found that clustered nuclei encompass at least two phenotypes that are caused by distinct mechanisms. Specifically, Ensconsin is necessary for productive force production to drive any movement of nuclei, whereas Bocksbeutel and Klarsicht are necessary to form distinct populations of nuclei that move to different cellular locations. Mechanistically, Ensconsin regulates the number of growing microtubules that are used to move nuclei, whereas Bocksbeutel and Klarsicht regulate interactions between nuclei.
Collapse
Affiliation(s)
| | - L Alexis Coon
- Department of Biology, Boston College, Chestnut Hill, MA 02467
| | - Riya Thomas
- Department of Biology, Boston College, Chestnut Hill, MA 02467
| | | | - Elizabeth Wynn
- Department of Biology, Boston College, Chestnut Hill, MA 02467
| | - Eric S Folker
- Department of Biology, Boston College, Chestnut Hill, MA 02467
| |
Collapse
|
28
|
Denarier E, Ecklund KH, Berthier G, Favier A, O'Toole ET, Gory-Fauré S, De Macedo L, Delphin C, Andrieux A, Markus SM, Boscheron C. Modeling a disease-correlated tubulin mutation in budding yeast reveals insight into MAP-mediated dynein function. Mol Biol Cell 2021; 32:ar10. [PMID: 34379441 PMCID: PMC8684761 DOI: 10.1091/mbc.e21-05-0237] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Mutations in the genes that encode α- and β-tubulin underlie many neurological diseases, most notably malformations in cortical development. In addition to revealing the molecular basis for disease etiology, studying such mutations can provide insight into microtubule function and the role of the large family of microtubule effectors. In this study, we use budding yeast to model one such mutation—Gly436Arg in α-tubulin, which is causative of malformations in cortical development—in order to understand how it impacts microtubule function in a simple eukaryotic system. Using a combination of in vitro and in vivo methodologies, including live cell imaging and electron tomography, we find that the mutant tubulin is incorporated into microtubules, causes a shift in α-tubulin isotype usage, and dramatically enhances dynein activity, which leads to spindle-positioning defects. We find that the basis for the latter phenotype is an impaired interaction between She1—a dynein inhibitor—and the mutant microtubules. In addition to revealing the natural balance of α-tubulin isotype utilization in cells, our results provide evidence of an impaired interaction between microtubules and a dynein regulator as a consequence of a tubulin mutation and sheds light on a mechanism that may be causative of neurodevelopmental diseases.
Collapse
Affiliation(s)
- E Denarier
- Univ. Grenoble Alpes, CEA, CNRS, GIN, IBS, Inserm, IRIG, F-38000 Grenoble, France
| | - K H Ecklund
- Department of Biochemistry and Molecular Biology, Colorado State University, Fort Collins, Colorado, United States
| | - G Berthier
- Univ. Grenoble Alpes, CEA, CNRS, GIN, IBS, Inserm, IRIG, F-38000 Grenoble, France
| | - A Favier
- Univ. Grenoble Alpes, CEA, CNRS, GIN, IBS, Inserm, IRIG, F-38000 Grenoble, France
| | - E T O'Toole
- Department of Molecular, Cellular, and Developmental Biology, University of Colorado, Boulder, Boulder, Colorado, United States
| | - S Gory-Fauré
- Univ. Grenoble Alpes, CEA, CNRS, GIN, IBS, Inserm, IRIG, F-38000 Grenoble, France
| | - L De Macedo
- Univ. Grenoble Alpes, CEA, CNRS, GIN, IBS, Inserm, IRIG, F-38000 Grenoble, France
| | - C Delphin
- Univ. Grenoble Alpes, CEA, CNRS, GIN, IBS, Inserm, IRIG, F-38000 Grenoble, France
| | - A Andrieux
- Univ. Grenoble Alpes, CEA, CNRS, GIN, IBS, Inserm, IRIG, F-38000 Grenoble, France
| | - S M Markus
- Department of Biochemistry and Molecular Biology, Colorado State University, Fort Collins, Colorado, United States
| | - C Boscheron
- Univ. Grenoble Alpes, CEA, CNRS, GIN, IBS, Inserm, IRIG, F-38000 Grenoble, France
| |
Collapse
|
29
|
Qian C, Dong B, Wang XY, Zhou FQ. In vivo glial trans-differentiation for neuronal replacement and functional recovery in central nervous system. FEBS J 2021; 288:4773-4785. [PMID: 33351267 PMCID: PMC8217397 DOI: 10.1111/febs.15681] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Revised: 12/04/2020] [Accepted: 12/21/2020] [Indexed: 02/07/2023]
Abstract
The adult mammalian central nervous system (CNS) is deficient in intrinsic machineries to replace neurons lost in injuries or progressive degeneration. Various types of these neurons constitute neural circuitries wired to support vital sensory, motor, and cognitive functions. Based on the pioneer studies in cell lineage conversion, one promising strategy is to convert in vivo glial cells into neural progenitors or directly into neurons that can be eventually rewired for functional recovery. We first briefly summarize the well-studied regeneration-capable CNS in the zebrafish, focusing on their postinjury spontaneous reprogramming of the retinal Müller glia (MG). We then compare the signaling transductions, and transcriptional and epigenetic regulations in the zebrafish MGs with their mammalian counterparts, which perpetuate certain barriers against proliferation and neurogenesis and thus fail in MG-to-progenitor conversion. Next, we discuss emerging evidence from mouse studies, in which the in vivo glia-to-neuron conversion could be achieved with sequential or one-step genetic manipulations, such as the conversions from retinal MGs to interneurons, photoreceptors, or retinal ganglion cells (RGCs), as well as the conversions from midbrain astrocytes to dopaminergic or GABAergic neurons. Some of these in vivo studies showed considerable coverage of subtypes in the newly induced neurons and partial reestablishment in neural circuits and functions. Importantly, we would like to point out some crucial technical concerns that need to be addressed to convincingly show successful glia-to-neuron conversion. Finally, we present challenges and future directions in the field for better neural function recovery.
Collapse
Affiliation(s)
- Cheng Qian
- Department of Orthopaedic Surgery, The Johns Hopkins University School of Medicine, Baltimore, MD 21287
| | - Bryan Dong
- Department of Orthopaedic Surgery, The Johns Hopkins University School of Medicine, Baltimore, MD 21287
| | - Xu-Yang Wang
- Department of Orthopaedic Surgery, The Johns Hopkins University School of Medicine, Baltimore, MD 21287
| | - Feng-Quan Zhou
- Department of Orthopaedic Surgery, The Johns Hopkins University School of Medicine, Baltimore, MD 21287,The Solomon H. Snyder Department of Neuroscience, The Johns Hopkins University School of Medicine, Baltimore, MD 21287,Corresponding author: Feng-Quan Zhou, Ph.D., , Phone: 443-287-5649, Address: The John G. Rangos Building, Room 291, 855 North Wolfe Street, Baltimore, MD 21205, USA
| |
Collapse
|
30
|
Jiang M, Tang T, Liang X, Li J, Qiu Y, Liu S, Bian S, Xie Y, Fang F, Cang J. Maternal sevoflurane exposure induces temporary defects in interkinetic nuclear migration of radial glial progenitors in the fetal cerebral cortex through the Notch signalling pathway. Cell Prolif 2021; 54:e13042. [PMID: 33955094 PMCID: PMC8168415 DOI: 10.1111/cpr.13042] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Revised: 03/02/2021] [Accepted: 04/03/2021] [Indexed: 12/13/2022] Open
Abstract
OBJECTIVES The effects of general anaesthetics on fetal brain development remain elusive. Radial glial progenitors (RGPs) generate the majority of neurons in developing brains. Here, we evaluated the acute alterations in RGPs after maternal sevoflurane exposure. METHODS Pregnant mice were exposed to 2.5% sevoflurane for 6 hours on gestational day 14.5. Interkinetic nuclear migration (INM) of RGPs in the ventricular zone (VZ) of the fetal brain was evaluated by thymidine analogues labelling. Cell fate of RGP progeny was determined by immunostaining using various neural markers. The Morris water maze (MWM) was used to assess the neurocognitive behaviours of the offspring. RNA sequencing (RNA-Seq) was performed for the potential mechanism, and the potential mechanism validated by quantitative real-time PCR (qPCR), Western blot and rescue experiments. Furthermore, INM was examined in human embryonic stem cell (hESC)-derived 3D cerebral organoids. RESULTS Maternal sevoflurane exposure induced temporary abnormities in INM, and disturbed the cell cycle progression of RGPs in both rodents and cerebral organoids without cell fate alternation. RNA-Seq analysis, qPCR and Western blot showed that the Notch signalling pathway was a potential downstream target. Reactivation of Notch by Jag1 and NICD overexpression rescued the defects in INM. Young adult offspring showed no obvious cognitive impairments in MWM. CONCLUSIONS Maternal sevoflurane exposure during neurogenic period temporarily induced abnormal INM of RGPs by targeting the Notch signalling pathway without inducing long-term effects on RGP progeny cell fate or offspring cognitive behaviours. More importantly, the defects of INM in hESC-derived cerebral organoids provide a novel insight into the effects of general anaesthesia on human brain development.
Collapse
Affiliation(s)
- Ming Jiang
- Department of AnesthesiaZhongshan HospitalFudan UniversityShanghaiChina
| | - Tianxiang Tang
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain ScienceInstitutes of Brain ScienceFudan UniversityShanghaiChina
| | - Xinyue Liang
- Department of AnesthesiaZhongshan HospitalFudan UniversityShanghaiChina
| | - Juchen Li
- Department of AnesthesiaZhongshan HospitalFudan UniversityShanghaiChina
| | - Yue Qiu
- Department of AnesthesiaZhongshan HospitalFudan UniversityShanghaiChina
| | - Shiwen Liu
- Department of AnesthesiaZhongshan HospitalFudan UniversityShanghaiChina
| | - Shan Bian
- Institute for Regenerative MedicineSchool of Life Sciences and TechnologyFrontier Science Center for Stem Cell ResearchShanghai East HospitalTongji UniversityShanghaiChina
| | - Yunli Xie
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain ScienceInstitutes of Brain ScienceFudan UniversityShanghaiChina
| | - Fang Fang
- Department of AnesthesiaZhongshan HospitalFudan UniversityShanghaiChina
| | - Jing Cang
- Department of AnesthesiaZhongshan HospitalFudan UniversityShanghaiChina
| |
Collapse
|
31
|
Getachew D, Matsumoto A, Uchimura Y, Udagawa J, Mita N, Ogawa N, Moriyama S, Takami A, Otani H. Global pattern of interkinetic nuclear migration in tracheoesophageal epithelia of the mouse embryo: Interorgan and intraorgan regional differences. Congenit Anom (Kyoto) 2021; 61:82-96. [PMID: 33249638 DOI: 10.1111/cga.12405] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Revised: 11/13/2020] [Accepted: 11/23/2020] [Indexed: 12/01/2022]
Abstract
Interkinetic nuclear migration (INM) is an apicobasal (AB) polarity-based regulatory mechanism of proliferation/differentiation in epithelial stem/progenitor cells. We previously documented INM in the endoderm-derived tracheal/esophageal epithelia at embryonic day (E) 11.5 and suggested that INM is involved in the development of both organs. We here investigated interorgan (trachea vs esophagus) and intraorgan regional (ventral vs dorsal) differences in the INM mode in the tracheal and esophageal epithelia of the mouse embryo. We also analyzed convergent extension (CE) and planar cell movement (PCM) in the epithelia based on cell distribution. The pregnant C57BL/6J mice were intraperitoneally injected with 5-ethynyl-2'-deoxyuridine at E11.5 and E12.5 and were sacrificed 1, 4, 6, 8, and 12 hours later to obtain the embryos. The distribution of labeled cell nuclei along the AB axis was chronologically analyzed in the total, ventral, and dorsal sides of the epithelia. The percentage distribution of the nuclei population was represented by histogram and the chronological change was analyzed statistically using multidimensional scaling. The interorgan comparison of the INM mode during E11.5-E12.0, but not E12.5-E13.0, showed a significant difference. During E11.5-E12.0 the trachea, but not the esophagus, showed a significant difference between ventral and dorsal sides. During E12.5-E13.0 neither organ showed regional differences. CE appeared to occur in both organs during E11.5-E12.0 while PCM was unclear in both organs. These findings suggest a difference between the trachea and esophagus, and a regional difference in the trachea, not in the esophagus, in the INM mode, which may be related with the later differential organogenesis/histogenesis of these organs.
Collapse
Affiliation(s)
- Dereje Getachew
- Department of Developmental Biology, Faculty of Medicine, Shimane University, Izumo, Japan
| | - Akihiro Matsumoto
- Department of Developmental Biology, Faculty of Medicine, Shimane University, Izumo, Japan
| | - Yasuhiro Uchimura
- Department of Anatomy, Shiga University of Medical Science, Otsu, Japan
| | - Jun Udagawa
- Department of Anatomy, Shiga University of Medical Science, Otsu, Japan
| | - Nanako Mita
- Department of Developmental Biology, Faculty of Medicine, Shimane University, Izumo, Japan
| | - Noriko Ogawa
- Department of Developmental Biology, Faculty of Medicine, Shimane University, Izumo, Japan
| | - Shigeru Moriyama
- Department of Developmental Biology, Faculty of Medicine, Shimane University, Izumo, Japan
| | - Akiyasu Takami
- Department of Mechanical Engineering, National Institute of Technology, Matsue College, Matsue, Japan
| | - Hiroki Otani
- Department of Developmental Biology, Faculty of Medicine, Shimane University, Izumo, Japan
| |
Collapse
|
32
|
Shin D, Nakamura M, Morishita Y, Eiraku M, Yamakawa T, Sasamura T, Akiyama M, Inaki M, Matsuno K. Collective nuclear behavior shapes bilateral nuclear symmetry for subsequent left-right asymmetric morphogenesis in Drosophila. Development 2021; 148:260539. [PMID: 34097729 PMCID: PMC8126412 DOI: 10.1242/dev.198507] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Accepted: 03/23/2021] [Indexed: 11/20/2022]
Abstract
Proper organ development often requires nuclei to move to a specific position within the cell. To determine how nuclear positioning affects left-right (LR) development in the Drosophila anterior midgut (AMG), we developed a surface-modeling method to measure and describe nuclear behavior at stages 13-14, captured in three-dimensional time-lapse movies. We describe the distinctive positioning and a novel collective nuclear behavior by which nuclei align LR symmetrically along the anterior-posterior axis in the visceral muscles that overlie the midgut and are responsible for the LR-asymmetric development of this organ. Wnt4 signaling is crucial for the collective behavior and proper positioning of the nuclei, as are myosin II and the LINC complex, without which the nuclei fail to align LR symmetrically. The LR-symmetric positioning of the nuclei is important for the subsequent LR-asymmetric development of the AMG. We propose that the bilaterally symmetrical positioning of these nuclei may be mechanically coupled with subsequent LR-asymmetric morphogenesis.
Collapse
Affiliation(s)
- Dongsun Shin
- Department of Biological Sciences, Osaka University, 1-1 Machikaneyama, Toyonaka, Osaka 560-0043, Japan
| | - Mitsutoshi Nakamura
- Department of Biological Sciences, Osaka University, 1-1 Machikaneyama, Toyonaka, Osaka 560-0043, Japan
| | - Yoshitaka Morishita
- Department of Biological Sciences, Osaka University, 1-1 Machikaneyama, Toyonaka, Osaka 560-0043, Japan
| | - Mototsugu Eiraku
- Department of Biosystems Science, Kyoto University, 53 Shogoin Kawahara-cho, Sakyo-ku, Kyoto 606-8507, Japan
| | - Tomoko Yamakawa
- Department of Biological Sciences, Osaka University, 1-1 Machikaneyama, Toyonaka, Osaka 560-0043, Japan
| | - Takeshi Sasamura
- Department of Biological Sciences, Osaka University, 1-1 Machikaneyama, Toyonaka, Osaka 560-0043, Japan
| | - Masakazu Akiyama
- Meiji Institute for Advanced Study of Mathematical Sciences, Meiji University, 4-21-1 Nakano, Nakanoku, Tokyo 164-8525, Japan
| | - Mikiko Inaki
- Department of Biological Sciences, Osaka University, 1-1 Machikaneyama, Toyonaka, Osaka 560-0043, Japan
| | - Kenji Matsuno
- Department of Biological Sciences, Osaka University, 1-1 Machikaneyama, Toyonaka, Osaka 560-0043, Japan
| |
Collapse
|
33
|
Burger CA, Jiang D, Mackin RD, Samuel MA. Development and maintenance of vision's first synapse. Dev Biol 2021; 476:218-239. [PMID: 33848537 DOI: 10.1016/j.ydbio.2021.04.001] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Revised: 04/02/2021] [Accepted: 04/03/2021] [Indexed: 12/21/2022]
Abstract
Synapses in the outer retina are the first information relay points in vision. Here, photoreceptors form synapses onto two types of interneurons, bipolar cells and horizontal cells. Because outer retina synapses are particularly large and highly ordered, they have been a useful system for the discovery of mechanisms underlying synapse specificity and maintenance. Understanding these processes is critical to efforts aimed at restoring visual function through repairing or replacing neurons and promoting their connectivity. We review outer retina neuron synapse architecture, neural migration modes, and the cellular and molecular pathways that play key roles in the development and maintenance of these connections. We further discuss how these mechanisms may impact connectivity in the retina.
Collapse
Affiliation(s)
- Courtney A Burger
- Huffington Center on Aging, Department of Neuroscience, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Danye Jiang
- Huffington Center on Aging, Department of Neuroscience, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Robert D Mackin
- Huffington Center on Aging, Department of Neuroscience, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Melanie A Samuel
- Huffington Center on Aging, Department of Neuroscience, Baylor College of Medicine, Houston, TX, 77030, USA.
| |
Collapse
|
34
|
Wang M, Du L, Lee AC, Li Y, Qin H, He J. Different lineage contexts direct common pro-neural factors to specify distinct retinal cell subtypes. J Cell Biol 2021; 219:151968. [PMID: 32699896 PMCID: PMC7480095 DOI: 10.1083/jcb.202003026] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2020] [Revised: 05/13/2020] [Accepted: 06/04/2020] [Indexed: 02/08/2023] Open
Abstract
How astounding neuronal diversity arises from variable cell lineages in vertebrates remains mostly elusive. By in vivo lineage tracing of ∼1,000 single zebrafish retinal progenitors, we identified a repertoire of subtype-specific stereotyped neurogenic lineages. Remarkably, within these stereotyped lineages, GABAergic amacrine cells were born with photoreceptor cells, whereas glycinergic amacrine cells were born with OFF bipolar cells. More interestingly, post-mitotic differentiation blockage of GABAergic and glycinergic amacrine cells resulted in their respecification into photoreceptor and bipolar cells, respectively, suggesting lineage constraint in cell subtype specification. Using single-cell RNA-seq and ATAC-seq analyses, we further identified lineage-specific progenitors, each defined by specific transcription factors that exhibited characteristic chromatin accessibility dynamics. Finally, single pro-neural factors could specify different neuron types/subtypes in a lineage-dependent manner. Our findings reveal the importance of lineage context in defining neuronal subtypes and provide a demonstration of in vivo lineage-dependent induction of unique retinal neuron subtypes for treatment purposes.
Collapse
Affiliation(s)
- Mei Wang
- State Key Laboratory of Neuroscience, Institute of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China.,University of Chinese Academy of Sciences, Beijing, China.,Shanghai Center for Brain Science and Brain-Inspired Intelligence Technology, Shanghai, China
| | - Lei Du
- State Key Laboratory of Neuroscience, Institute of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China.,University of Chinese Academy of Sciences, Beijing, China.,Shanghai Center for Brain Science and Brain-Inspired Intelligence Technology, Shanghai, China
| | - Aih Cheun Lee
- State Key Laboratory of Neuroscience, Institute of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| | - Yan Li
- State Key Laboratory of Neuroscience, Institute of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China.,University of Chinese Academy of Sciences, Beijing, China.,Shanghai Center for Brain Science and Brain-Inspired Intelligence Technology, Shanghai, China
| | - Huiwen Qin
- State Key Laboratory of Neuroscience, Institute of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China.,University of Chinese Academy of Sciences, Beijing, China.,Shanghai Center for Brain Science and Brain-Inspired Intelligence Technology, Shanghai, China
| | - Jie He
- State Key Laboratory of Neuroscience, Institute of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China.,Shanghai Center for Brain Science and Brain-Inspired Intelligence Technology, Shanghai, China
| |
Collapse
|
35
|
Clark BS, Miesfeld JB, Flinn MA, Collery RF, Link BA. Dynamic Polarization of Rab11a Modulates Crb2a Localization and Impacts Signaling to Regulate Retinal Neurogenesis. Front Cell Dev Biol 2021; 8:608112. [PMID: 33634099 PMCID: PMC7900515 DOI: 10.3389/fcell.2020.608112] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2020] [Accepted: 12/28/2020] [Indexed: 01/31/2023] Open
Abstract
Interkinetic nuclear migration (IKNM) is the process in which pseudostratified epithelial nuclei oscillate from the apical to basal surface and in phase with the mitotic cycle. In the zebrafish retina, neuroepithelial retinal progenitor cells (RPCs) increase Notch activity with apical movement of the nuclei, and the depth of nuclear migration correlates with the probability that the next cell division will be neurogenic. This study focuses on the mechanisms underlying the relationships between IKNM, cell signaling, and neurogenesis. In particular, we have explored the role IKNM has on endosome biology within RPCs. Through genetic manipulation and live imaging in zebrafish, we find that early (Rab5-positive) and recycling (Rab11a-positive) endosomes polarize in a dynamic fashion within RPCs and with reference to nuclear position. Functional analyses suggest that dynamic polarization of recycling endosomes and their activity within the neuroepithelia modulates the subcellular localization of Crb2a, consequently affecting multiple signaling pathways that impact neurogenesis including Notch, Hippo, and Wnt activities. As nuclear migration is heterogenous and asynchronous among RPCs, Rab11a-affected signaling within the neuroepithelia is modulated in a differential manner, providing mechanistic insight to the correlation of IKNM and selection of RPCs to undergo neurogenesis.
Collapse
Affiliation(s)
- Brian S Clark
- Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Joel B Miesfeld
- Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Michael A Flinn
- Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, WI, United States.,Department of Physiology, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Ross F Collery
- Department of Ophthalmology and Visual Sciences, Medical College of Wisconsin Eye Institute, Milwaukee, WI, United States
| | - Brian A Link
- Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, WI, United States
| |
Collapse
|
36
|
Moser M, Kirkpatrick A, Groves NR, Meier I. LINC-complex mediated positioning of the vegetative nucleus is involved in calcium and ROS signaling in Arabidopsis pollen tubes. Nucleus 2020; 11:149-163. [PMID: 32631106 PMCID: PMC7529407 DOI: 10.1080/19491034.2020.1783783] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Nuclear movement and positioning play a role in developmental processes throughout life. Nuclear movement and positioning are mediated primarily by linker of nucleoskeleton and cytoskeleton (LINC) complexes. LINC complexes are comprised of the inner nuclear membrane SUN proteins and the outer nuclear membrane (ONM) KASH proteins. In Arabidopsis pollen tubes, the vegetative nucleus (VN) maintains a fixed distance from the pollen tube tip during growth, and the VN precedes the sperm cells (SCs). In pollen tubes of wit12 and wifi, mutants deficient in the ONM component of a plant LINC complex, the SCs precede the VN during pollen tube growth and the fixed VN distance from the tip is lost. Subsequently, pollen tubes frequently fail to burst upon reception. In this study, we sought to determine if the pollen tube reception defect observed in wit12 and wifi is due to decreased sensitivity to reactive oxygen species (ROS). Here, we show that wit12 and wifi are hyposensitive to exogenous H2O2, and that this hyposensitivity is correlated with decreased proximity of the VN to the pollen tube tip. Additionally, we report the first instance of nuclear Ca2+ peaks in growing pollen tubes, which are disrupted in the wit12 mutant. In the wit12 mutant, nuclear Ca2+ peaks are reduced in response to exogenous ROS, but these peaks are not correlated with pollen tube burst. This study finds that VN proximity to the pollen tube tip is required for both response to exogenous ROS, as well as internal nuclear Ca2+ fluctuations.
Collapse
Affiliation(s)
- Morgan Moser
- Department of Molecular Genetics, The Ohio State University , Columbus, OH, USA
| | - Andrew Kirkpatrick
- Department of Molecular Genetics, The Ohio State University , Columbus, OH, USA
| | - Norman Reid Groves
- Department of Molecular Genetics, The Ohio State University , Columbus, OH, USA.,Center for Applied Plant Sciences, The Ohio State University , Columbus, OH, USA
| | - Iris Meier
- Department of Molecular Genetics, The Ohio State University , Columbus, OH, USA.,Center for Applied Plant Sciences, The Ohio State University , Columbus, OH, USA.,Center for RNA Biology, The Ohio State University , Columbus, OH, USA
| |
Collapse
|
37
|
Retrograde Mitochondrial Transport Is Essential for Organelle Distribution and Health in Zebrafish Neurons. J Neurosci 2020; 41:1371-1392. [PMID: 33376159 PMCID: PMC7896009 DOI: 10.1523/jneurosci.1316-20.2020] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Revised: 11/25/2020] [Accepted: 12/01/2020] [Indexed: 12/12/2022] Open
Abstract
In neurons, mitochondria are transported by molecular motors throughout the cell to form and maintain functional neural connections. These organelles have many critical functions in neurons and are of high interest as their dysfunction is associated with disease. While the mechanics and impact of anterograde mitochondrial movement toward axon terminals are beginning to be understood, the frequency and function of retrograde (cell body directed) mitochondrial transport in neurons are still largely unexplored. While existing evidence indicates that some mitochondria are retrogradely transported for degradation in the cell body, the precise impact of disrupting retrograde transport on the organelles and the axon was unknown. Using long-term, in vivo imaging, we examined mitochondrial motility in zebrafish sensory and motor axons. We show that retrograde transport of mitochondria from axon terminals allows replacement of the axon terminal population within a day. By tracking these organelles, we show that not all mitochondria that leave the axon terminal are degraded; rather, they persist over several days. Disrupting retrograde mitochondrial flux in neurons leads to accumulation of aged organelles in axon terminals and loss of cell body mitochondria. Assays of neural circuit activity demonstrated that disrupting mitochondrial transport and function has no effect on sensory axon terminal activity but does negatively impact motor neuron axons. Taken together, our work supports a previously unappreciated role for retrograde mitochondrial transport in the maintenance of a homeostatic distribution of mitochondria in neurons and illustrates the downstream effects of disrupting this process on sensory and motor circuits. SIGNIFICANCE STATEMENT Disrupted mitochondrial transport has been linked to neurodegenerative disease. Retrograde transport of this organelle has been implicated in turnover of aged organelles through lysosomal degradation in the cell body. Consistent with this, we provide evidence that retrograde mitochondrial transport is important for removing aged organelles from axons; however, we show that these organelles are not solely degraded, rather they persist in neurons for days. Disrupting retrograde mitochondrial transport impacts the homeostatic distribution of mitochondria throughout the neuron and the function of motor, but not sensory, axon synapses. Together, our work shows the conserved reliance on retrograde mitochondrial transport for maintaining a healthy mitochondrial pool in neurons and illustrates the disparate effects of disrupting this process on sensory versus motor circuits.
Collapse
|
38
|
Soto-Perez J, Baumgartner M, Kanadia RN. Role of NDE1 in the Development and Evolution of the Gyrified Cortex. Front Neurosci 2020; 14:617513. [PMID: 33390896 PMCID: PMC7775536 DOI: 10.3389/fnins.2020.617513] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Accepted: 11/12/2020] [Indexed: 12/17/2022] Open
Abstract
An expanded cortex is a hallmark of human neurodevelopment and endows increased cognitive capabilities. Recent work has shown that the cell cycle-related gene NDE1 is essential for proper cortical development. Patients who have mutations in NDE1 exhibit congenital microcephaly as a primary phenotype. At the cellular level, NDE1 is essential for interkinetic nuclear migration and mitosis of radial glial cells, which translates to an indispensable role in neurodevelopment. The nuclear migration function of NDE1 is well conserved across Opisthokonta. In mammals, multiple isoforms containing alternate terminal exons, which influence the functionality of NDE1, have been reported. It has been noted that the pattern of terminal exon usage mirrors patterns of cortical complexity in mammals. To provide context to these findings, here, we provide a comprehensive review of the literature regarding NDE1, its molecular biology and physiological relevance at the cellular and organismal levels. In particular, we outline the potential roles of NDE1 in progenitor cell behavior and explore the spectrum of NDE1 pathogenic variants. Moreover, we assessed the evolutionary conservation of NDE1 and interrogated whether the usage of alternative terminal exons is characteristic of species with gyrencephalic cortices. We found that gyrencephalic species are more likely to express transcripts that use the human-associated terminal exon, whereas lissencephalic species tend to express transcripts that use the mouse-associated terminal exon. Among gyrencephalic species, the human-associated terminal exon was preferentially expressed by those with a high order of gyrification. These findings underscore phylogenetic relationships between the preferential usage of NDE1 terminal exon and high-order gyrification, which provide insight into cortical evolution underlying high-order brain functions.
Collapse
Affiliation(s)
- Jaseph Soto-Perez
- Department of Physiology and Neurobiology, University of Connecticut, Storrs, CT, United States
| | | | - Rahul N. Kanadia
- Department of Physiology and Neurobiology, University of Connecticut, Storrs, CT, United States
- Institute for Systems Genomics, University of Connecticut, Storrs, CT, United States
| |
Collapse
|
39
|
Pushchina EV, Stukaneva ME, Varaksin AA. Hydrogen Sulfide Modulates Adult and Reparative Neurogenesis in the Cerebellum of Juvenile Masu Salmon, Oncorhynchus masou. Int J Mol Sci 2020; 21:ijms21249638. [PMID: 33348868 PMCID: PMC7766854 DOI: 10.3390/ijms21249638] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Revised: 12/03/2020] [Accepted: 12/15/2020] [Indexed: 01/31/2023] Open
Abstract
Fish are a convenient model for the study of reparative and post-traumatic processes of central nervous system (CNS) recovery, because the formation of new cells in their CNS continues throughout life. After a traumatic injury to the cerebellum of juvenile masu salmon, Oncorhynchus masou, the cell composition of the neurogenic zones containing neural stem cells (NSCs)/neural progenitor cells (NPCs) in the acute period (two days post-injury) changes. The presence of neuroepithelial (NE) and radial glial (RG) neuronal precursors located in the dorsal, lateral, and basal zones of the cerebellar body was shown by the immunohistochemical (IHC) labeling of glutamine synthetase (GS). Progenitors of both types are sources of neurons in the cerebellum of juvenile O. masou during constitutive growth, thus, playing an important role in CNS homeostasis and neuronal plasticity during ontogenesis. Precursors with the RG phenotype were found in the same regions of the molecular layer as part of heterogeneous constitutive neurogenic niches. The presence of neuroepithelial and radial glia GS+ cells indicates a certain proportion of embryonic and adult progenitors and, obviously, different contributions of these cells to constitutive and reparative neurogenesis in the acute post-traumatic period. Expression of nestin and vimentin was revealed in neuroepithelial cerebellar progenitors of juvenile O. masou. Patterns of granular expression of these markers were found in neurogenic niches and adjacent areas, which probably indicates the neurotrophic and proneurogenic effects of vimentin and nestin in constitutive and post-traumatic neurogenesis and a high level of constructive metabolism. No expression of vimentin and nestin was detected in the cerebellar RG of juvenile O. masou. Thus, the molecular markers of NSCs/NPCs in the cerebellum of juvenile O. masou are as follows: vimentin, nestin, and glutamine synthetase label NE cells in intact animals and in the post-traumatic period, while GS expression is present in the RG of intact animals and decreases in the acute post-traumatic period. A study of distribution of cystathionine β-synthase (CBS) in the cerebellum of intact young O. masou showed the expression of the marker mainly in type 1 cells, corresponding to NSCs/NCPs for other molecular markers. In the post-traumatic period, the number of CBS+ cells sharply increased, which indicates the involvement of H2S in the post-traumatic response. Induction of CBS in type 3 cells indicates the involvement of H2S in the metabolism of extracellular glutamate in the cerebellum, a decrease in the production of reactive oxygen species, and also arrest of the oxidative stress development, a weakening of the toxic effects of glutamate, and a reduction in excitotoxicity. The obtained results allow us to consider H2S as a biologically active substance, the numerous known effects of which can be supplemented by participation in the processes of constitutive neurogenesis and neuronal regeneration.
Collapse
|
40
|
Nerli E, Rocha-Martins M, Norden C. Asymmetric neurogenic commitment of retinal progenitors involves Notch through the endocytic pathway. eLife 2020; 9:e60462. [PMID: 33141024 PMCID: PMC7679139 DOI: 10.7554/elife.60462] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Accepted: 11/02/2020] [Indexed: 01/07/2023] Open
Abstract
During brain development, progenitor cells need to balanceproliferation and differentiation in order to generate different neurons in the correct numbers and proportions. Currently, the patterns of multipotent progenitor divisions that lead to neurogenic entry and the factors that regulate them are not fully understood. We here use the zebrafish retina to address this gap, exploiting its suitability for quantitative live-imaging. We show that early neurogenic progenitors arise from asymmetric divisions. Notch regulates this asymmetry, as when inhibited, symmetric divisions producing two neurogenic progenitors occur. Surprisingly however, Notch does not act through an apicobasal activity gradient as previously suggested, but through asymmetric inheritance of Sara-positive endosomes. Further, the resulting neurogenic progenitors show cell biological features different from multipotent progenitors, raising the possibility that an intermediate progenitor state exists in the retina. Our study thus reveals new insights into the regulation of proliferative and differentiative events during central nervous system development.
Collapse
Affiliation(s)
- Elisa Nerli
- Max Planck Institute of Molecular Cell Biology and GeneticsDresdenGermany
- Instituto Gulbenkian de CiênciaOeirasPortugal
| | - Mauricio Rocha-Martins
- Max Planck Institute of Molecular Cell Biology and GeneticsDresdenGermany
- Instituto Gulbenkian de CiênciaOeirasPortugal
| | - Caren Norden
- Max Planck Institute of Molecular Cell Biology and GeneticsDresdenGermany
- Instituto Gulbenkian de CiênciaOeirasPortugal
| |
Collapse
|
41
|
Fourel G, Boscheron C. Tubulin mutations in neurodevelopmental disorders as a tool to decipher microtubule function. FEBS Lett 2020; 594:3409-3438. [PMID: 33064843 DOI: 10.1002/1873-3468.13958] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Revised: 09/28/2020] [Accepted: 10/05/2020] [Indexed: 01/08/2023]
Abstract
Malformations of cortical development (MCDs) are a group of severe brain malformations associated with intellectual disability and refractory childhood epilepsy. Human missense heterozygous mutations in the 9 α-tubulin and 10 β-tubulin isoforms forming the heterodimers that assemble into microtubules (MTs) were found to cause MCDs. However, how a single mutated residue in a given tubulin isoform can perturb the entire microtubule population in a neuronal cell remains a crucial question. Here, we examined 85 MCD-associated tubulin mutations occurring in TUBA1A, TUBB2, and TUBB3 and their location in a three-dimensional (3D) microtubule cylinder. Mutations hitting residues exposed on the outer microtubule surface are likely to alter microtubule association with partners, while alteration of intradimer contacts may impair dimer stability and straightness. Other types of mutations are predicted to alter interdimer and lateral contacts, which are responsible for microtubule cohesion, rigidity, and dynamics. MCD-associated tubulin mutations surprisingly fall into all categories, thus providing unexpected insights into how a single mutation may impair microtubule function and elicit dominant effects in neurons.
Collapse
|
42
|
Azizi A, Herrmann A, Wan Y, Buse SJ, Keller PJ, Goldstein RE, Harris WA. Nuclear crowding and nonlinear diffusion during interkinetic nuclear migration in the zebrafish retina. eLife 2020; 9:58635. [PMID: 33021471 PMCID: PMC7538155 DOI: 10.7554/elife.58635] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Accepted: 09/03/2020] [Indexed: 12/26/2022] Open
Abstract
An important question in early neural development is the origin of stochastic nuclear movement between apical and basal surfaces of neuroepithelia during interkinetic nuclear migration. Tracking of nuclear subpopulations has shown evidence of diffusion - mean squared displacements growing linearly in time - and suggested crowding from cell division at the apical surface drives basalward motion. Yet, this hypothesis has not yet been tested, and the forces involved not quantified. We employ long-term, rapid light-sheet and two-photon imaging of early zebrafish retinogenesis to track entire populations of nuclei within the tissue. The time-varying concentration profiles show clear evidence of crowding as nuclei reach close-packing and are quantitatively described by a nonlinear diffusion model. Considerations of nuclear motion constrained inside the enveloping cell membrane show that concentration-dependent stochastic forces inside cells, compatible in magnitude to those found in cytoskeletal transport, can explain the observed magnitude of the diffusion constant.
Collapse
Affiliation(s)
- Afnan Azizi
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, United Kingdom
| | - Anne Herrmann
- Department of Applied Mathematics and Theoretical Physics, Centre for Mathematical Sciences, University of Cambridge, Cambridge, United Kingdom
| | - Yinan Wan
- Howard Hughes Medical Institute, Janelia Research Campus, Ashburn, United States
| | - Salvador Jrp Buse
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, United Kingdom
| | - Philipp J Keller
- Howard Hughes Medical Institute, Janelia Research Campus, Ashburn, United States
| | - Raymond E Goldstein
- Department of Applied Mathematics and Theoretical Physics, Centre for Mathematical Sciences, University of Cambridge, Cambridge, United Kingdom
| | - William A Harris
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
43
|
Campbell LJ, Hobgood JS, Jia M, Boyd P, Hipp RI, Hyde DR. Notch3 and DeltaB maintain Müller glia quiescence and act as negative regulators of regeneration in the light-damaged zebrafish retina. Glia 2020; 69:546-566. [PMID: 32965734 DOI: 10.1002/glia.23912] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2019] [Revised: 09/04/2020] [Accepted: 09/07/2020] [Indexed: 12/13/2022]
Abstract
Damage to the zebrafish retina stimulates resident Müller glia to reprogram, reenter the cell cycle, divide asymmetrically, and produce neuronal progenitor cells that amplify and differentiate into the lost neurons. The transition from quiescent to proliferative Müller glia involves both positive and negative regulators. We previously demonstrated that the Notch signaling pathway represses retinal regeneration by maintaining Müller glia quiescence in zebrafish. Here we examine which Notch receptor is necessary to maintain quiescence. Quantitative RT-PCR and RNA-Seq analyses reveal that notch3 is expressed in the undamaged retina and is downregulated in response to light damage. Additionally, Notch3 protein is expressed in quiescent Müller glia of the undamaged retina, is downregulated as Müller glia proliferate, and is reestablished in the Müller glia. Knockdown of Notch3 is sufficient to induce Müller glia proliferation in undamaged retinas and enhances proliferation during light damage. Alternatively, knockdown of Notch1a, Notch1b, or Notch2 decreases the number of proliferating cells during light damage, suggesting that Notch signaling is also required for proliferation during retinal regeneration. We also knockdown the zebrafish Delta and Delta-like proteins, ligands for the Notch receptors, and find that the deltaB morphant possesses an increased number of proliferating cells in the light-damaged retina. As with Notch3, knockdown of DeltaB is sufficient to induce Müller glia proliferation in the absence of light damage. Taken together, the negative regulation of Müller glia proliferation in zebrafish retinal regeneration is mediated by Notch3 and DeltaB.
Collapse
Affiliation(s)
- Leah J Campbell
- Department of Biological Sciences, Center for Zebrafish Research, and the Center for Stem Cells and Regenerative Medicine, Galvin Life Science Center, University of Notre Dame, Notre Dame, Indiana, USA
| | - Joshua S Hobgood
- Department of Biological Sciences, Center for Zebrafish Research, and the Center for Stem Cells and Regenerative Medicine, Galvin Life Science Center, University of Notre Dame, Notre Dame, Indiana, USA
| | - Meng Jia
- Department of Biological Sciences, Center for Zebrafish Research, and the Center for Stem Cells and Regenerative Medicine, Galvin Life Science Center, University of Notre Dame, Notre Dame, Indiana, USA
| | - Patrick Boyd
- Department of Biological Sciences, Center for Zebrafish Research, and the Center for Stem Cells and Regenerative Medicine, Galvin Life Science Center, University of Notre Dame, Notre Dame, Indiana, USA
| | - Rebecca I Hipp
- Department of Biological Sciences, Center for Zebrafish Research, and the Center for Stem Cells and Regenerative Medicine, Galvin Life Science Center, University of Notre Dame, Notre Dame, Indiana, USA
| | - David R Hyde
- Department of Biological Sciences, Center for Zebrafish Research, and the Center for Stem Cells and Regenerative Medicine, Galvin Life Science Center, University of Notre Dame, Notre Dame, Indiana, USA
| |
Collapse
|
44
|
Zechner C, Nerli E, Norden C. Stochasticity and determinism in cell fate decisions. Development 2020; 147:147/14/dev181495. [PMID: 32669276 DOI: 10.1242/dev.181495] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
During development, cells need to make decisions about their fate in order to ensure that the correct numbers and types of cells are established at the correct time and place in the embryo. Such cell fate decisions are often classified as deterministic or stochastic. However, although these terms are clearly defined in a mathematical sense, they are sometimes used ambiguously in biological contexts. Here, we provide some suggestions on how to clarify the definitions and usage of the terms stochastic and deterministic in biological experiments. We discuss the frameworks within which such clear definitions make sense and highlight when certain ambiguity prevails. As an example, we examine how these terms are used in studies of neuronal cell fate decisions and point out areas in which definitions and interpretations have changed and matured over time. We hope that this Review will provide some clarification and inspire discussion on the use of terminology in relation to fate decisions.
Collapse
Affiliation(s)
- Christoph Zechner
- Max Planck Institute of Molecular Cell Biology and Genetics, Pfotenhauerstraße 108, 01307 Dresden, Germany .,Max Planck Center for Systems Biology, Pfotenhauerstraße 108, 01307 Dresden, Germany.,Cluster of Excellence Physics of Life, TU Dresden, 01062 Dresden, Germany
| | - Elisa Nerli
- Max Planck Institute of Molecular Cell Biology and Genetics, Pfotenhauerstraße 108, 01307 Dresden, Germany
| | - Caren Norden
- Max Planck Institute of Molecular Cell Biology and Genetics, Pfotenhauerstraße 108, 01307 Dresden, Germany .,Instituto Gulbenkian de Ciência, Rua da Quinta Grande 6, 2780-156 Oeiras, Portugal
| |
Collapse
|
45
|
Covello G, Rossello FJ, Filosi M, Gajardo F, Duchemin A, Tremonti BF, Eichenlaub M, Polo JM, Powell D, Ngai J, Allende ML, Domenici E, Ramialison M, Poggi L. Transcriptome analysis of the zebrafish atoh7-/- Mutant, lakritz, highlights Atoh7-dependent genetic networks with potential implications for human eye diseases. FASEB Bioadv 2020; 2:434-448. [PMID: 32676583 PMCID: PMC7354691 DOI: 10.1096/fba.2020-00030] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Revised: 04/02/2020] [Accepted: 04/28/2020] [Indexed: 12/21/2022] Open
Abstract
Expression of the bHLH transcription protein Atoh7 is a crucial factor conferring competence to retinal progenitor cells for the development of retinal ganglion cells. Several studies have emerged establishing ATOH7 as a retinal disease gene. Remarkably, such studies uncovered ATOH7 variants associated with global eye defects including optic nerve hypoplasia, microphthalmia, retinal vascular disorders, and glaucoma. The complex genetic networks and cellular decisions arising downstream of atoh7 expression, and how their dysregulation cause development of such disease traits remains unknown. To begin to understand such Atoh7-dependent events in vivo, we performed transcriptome analysis of wild-type and atoh7 mutant (lakritz) zebrafish embryos at the onset of retinal ganglion cell differentiation. We investigated in silico interplays of atoh7 and other disease-related genes and pathways. By network reconstruction analysis of differentially expressed genes, we identified gene clusters enriched in retinal development, cell cycle, chromatin remodeling, stress response, and Wnt pathways. By weighted gene coexpression network, we identified coexpression modules affected by the mutation and enriched in retina development genes tightly connected to atoh7. We established the groundwork whereby Atoh7-linked cellular and molecular processes can be investigated in the dynamic multi-tissue environment of the developing normal and diseased vertebrate eye.
Collapse
Affiliation(s)
- Giuseppina Covello
- Department of Cellular, Computational and Integrative Biology ‐ CIBIOUniversity of TrentoTrentoItaly
- Present address:
Department of BiologyUniversity of PadovaPadovaItaly
| | - Fernando J. Rossello
- Australian Regenerative Medicine InstituteMonash University Clayton VICClaytonAustralia
- Present address:
University of Melbourne Centre for Cancer ResearchUniversity of MelbourneMelbourneVictoriaAustralia
| | - Michele Filosi
- Department of Cellular, Computational and Integrative Biology ‐ CIBIOUniversity of TrentoTrentoItaly
| | - Felipe Gajardo
- Center for Genome RegulationFacultad de Ciencias, SantiagoUniversidad de ChileSantiagoChile
| | | | - Beatrice F. Tremonti
- Department of Cellular, Computational and Integrative Biology ‐ CIBIOUniversity of TrentoTrentoItaly
| | - Michael Eichenlaub
- Australian Regenerative Medicine InstituteMonash University Clayton VICClaytonAustralia
| | - Jose M. Polo
- Australian Regenerative Medicine InstituteMonash University Clayton VICClaytonAustralia
- BDIMonash University Clayton VICClaytonAustralia
| | - David Powell
- Monash Bioinformatics PlatformMonash University Clayton VICClaytonAustralia
| | - John Ngai
- Department of Molecular and Cell Biology & Helen Wills Neuroscience InstituteUniversity of CaliforniaBerkeleyCAUSA
| | - Miguel L. Allende
- Center for Genome RegulationFacultad de Ciencias, SantiagoUniversidad de ChileSantiagoChile
| | - Enrico Domenici
- Department of Cellular, Computational and Integrative Biology ‐ CIBIOUniversity of TrentoTrentoItaly
- Fondazione The Microsoft Research ‐ University of Trento Centre for Computational and Systems BiologyTrentoItaly
| | - Mirana Ramialison
- Australian Regenerative Medicine InstituteMonash University Clayton VICClaytonAustralia
| | - Lucia Poggi
- Department of Cellular, Computational and Integrative Biology ‐ CIBIOUniversity of TrentoTrentoItaly
- Centre for Organismal StudyHeidelberg UniversityHeidelbergGermany
- Department of PhysiologyDevelopment and NeuroscienceUniversity of CambridgeCambridgeUnited Kingdom
| |
Collapse
|
46
|
Moore R, Alexandre P. Delta-Notch Signaling: The Long and The Short of a Neuron's Influence on Progenitor Fates. J Dev Biol 2020; 8:jdb8020008. [PMID: 32225077 PMCID: PMC7345741 DOI: 10.3390/jdb8020008] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Revised: 03/20/2020] [Accepted: 03/24/2020] [Indexed: 01/16/2023] Open
Abstract
Maintenance of the neural progenitor pool during embryonic development is essential to promote growth of the central nervous system (CNS). The CNS is initially formed by tightly compacted proliferative neuroepithelial cells that later acquire radial glial characteristics and continue to divide at the ventricular (apical) and pial (basal) surface of the neuroepithelium to generate neurons. While neural progenitors such as neuroepithelial cells and apical radial glia form strong connections with their neighbours at the apical and basal surfaces of the neuroepithelium, neurons usually form the mantle layer at the basal surface. This review will discuss the existing evidence that supports a role for neurons, from early stages of differentiation, in promoting progenitor cell fates in the vertebrates CNS, maintaining tissue homeostasis and regulating spatiotemporal patterning of neuronal differentiation through Delta-Notch signalling.
Collapse
Affiliation(s)
- Rachel Moore
- Centre for Developmental Neurobiology, King’s College London, London SE1 1UL, UK
- Correspondence: (R.M.); (P.A.)
| | - Paula Alexandre
- Developmental Biology and Cancer, University College London Great Ormond Street Institute of Child Health, London WC1N 1EH, UK
- Correspondence: (R.M.); (P.A.)
| |
Collapse
|
47
|
The extracellular and intracellular regions of Crb2a play distinct roles in guiding the formation of the apical zonula adherens. Biomed Pharmacother 2020; 125:109942. [PMID: 32044715 DOI: 10.1016/j.biopha.2020.109942] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2019] [Revised: 01/18/2020] [Accepted: 01/23/2020] [Indexed: 11/22/2022] Open
Abstract
The transmembrane protein Crumbs (Crb), a key regulator of apical polarity, has a known involvement in establishment of the apical zonula adherens in epithelia, although the precise mechanism remains elusive. The zonula adherens are required to maintain the integrity and orderly arrangement of epithelia. Loss of the zonula adherens leads to morphogenetic defects in the tissues derived from epithelium. In this study, we revealed that the intracellular tail of Crb2a promoted the apical distribution of adherens junctions (AJs) in zebrafish retinal and lens epithelia, but caused assembly into unstable punctum adherens-like adhesion plaques. The extracellular region of Crb2a guided the transformation of AJs from the punctum adherens into stable zonula adherens. Accordingly, a truncated form of Crb2a lacking the extracellular region (Crb2aΔEX) could only partially rescue the retinal patterning defects in crb2a null mutant zebrafish (crb2am289). By contrast, constitutive over-expression of Crb2aΔEX disrupted the integrity of the outer limiting membrane in photoreceptors, which is derived from the zonula adherens of the retinal neuroepithelium. This study demonstrated that both the extracellular region and the intracellular tail of Crb2a are required to guide the formation of the apical zonula adherens.
Collapse
|
48
|
Veeraval L, O'Leary CJ, Cooper HM. Adherens Junctions: Guardians of Cortical Development. Front Cell Dev Biol 2020; 8:6. [PMID: 32117958 PMCID: PMC7025593 DOI: 10.3389/fcell.2020.00006] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Accepted: 01/10/2020] [Indexed: 12/01/2022] Open
Abstract
Apical radial glia comprise the pseudostratified neuroepithelium lining the embryonic lateral ventricles and give rise to the extensive repertoire of pyramidal neuronal subtypes of the neocortex. The establishment of a highly apicobasally polarized radial glial morphology is a mandatory prerequisite for cortical development as it governs neurogenesis, neural migration and the integrity of the ventricular wall. As in all epithelia, cadherin-based adherens junctions (AJs) play an obligate role in the maintenance of radial glial apicobasal polarity and neuroepithelial cohesion. In addition, the assembly of resilient AJs is critical to the integrity of the neuroepithelium which must resist the tensile forces arising from increasing CSF volume and other mechanical stresses associated with the expansion of the ventricles in the embryo and neonate. Junctional instability leads to the collapse of radial glial morphology, disruption of the ventricular surface and cortical lamination defects due to failed neuronal migration. The fidelity of cortical development is therefore dependent on AJ assembly and stability. Mutations in genes known to control radial glial junction formation are causative for a subset of inherited cortical malformations (neuronal heterotopias) as well as perinatal hydrocephalus, reinforcing the concept that radial glial junctions are pivotal determinants of successful corticogenesis. In this review we explore the key animal studies that have revealed important insights into the role of AJs in maintaining apical radial glial morphology and function, and as such, have provided a deeper understanding of the aberrant molecular and cellular processes contributing to debilitating cortical malformations. We highlight the reciprocal interactions between AJs and the epithelial polarity complexes that impose radial glial apicobasal polarity. We also discuss the critical molecular networks promoting AJ assembly in apical radial glia and emphasize the role of the actin cytoskeleton in the stabilization of cadherin adhesion – a crucial factor in buffering the mechanical forces exerted as a consequence of cortical expansion.
Collapse
Affiliation(s)
- Lenin Veeraval
- Queensland Brain Institute, The University of Queensland, Brisbane, QLD, Australia
| | - Conor J O'Leary
- Queensland Brain Institute, The University of Queensland, Brisbane, QLD, Australia
| | - Helen M Cooper
- Queensland Brain Institute, The University of Queensland, Brisbane, QLD, Australia
| |
Collapse
|
49
|
Vujovic F, Hunter N, Farahani RM. Notch pathway: a bistable inducer of biological noise? Cell Commun Signal 2019; 17:133. [PMID: 31640734 PMCID: PMC6805690 DOI: 10.1186/s12964-019-0453-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Accepted: 10/04/2019] [Indexed: 12/11/2022] Open
Abstract
Notch signalling pathway is central to development of metazoans. The pathway codes a binary fate switch. Upon activation, downstream signals contribute to resolution of fate dichotomies such as proliferation/differentiation or sub-lineage differentiation outcome. There is, however, an interesting paradox in the Notch signalling pathway. Despite remarkable predictability of fate outcomes instructed by the Notch pathway, the associated transcriptome is versatile and plastic. This inconsistency suggests the presence of an interface that compiles input from the plastic transcriptome of the Notch pathway but communicates only a binary output in biological decisions. Herein, we address the interface that determines fate outcomes. We provide an alternative hypothesis for the Notch pathway as a biological master switch that operates by induction of genetic noise and bistability in order to facilitate resolution of dichotomous fate outcomes in development.
Collapse
Affiliation(s)
- Filip Vujovic
- IDR/Westmead Institute for Medical Research, Sydney, Australia
- Faculty of Medicine and Health, University of Sydney, Sydney, NSW 2145 Australia
| | - Neil Hunter
- IDR/Westmead Institute for Medical Research, Sydney, Australia
- Faculty of Medicine and Health, University of Sydney, Sydney, NSW 2145 Australia
| | - Ramin M. Farahani
- IDR/Westmead Institute for Medical Research, Sydney, Australia
- Faculty of Medicine and Health, University of Sydney, Sydney, NSW 2145 Australia
| |
Collapse
|
50
|
Falk N, Joachimsthaler A, Kessler K, Lux UT, Noegel AA, Kremers J, Brandstätter JH, Gießl A, Falk N, Joachimsthaler A, Kessler K, Lux UT, Noegel AA, Kremers J, Brandstätter JH, Gießl A. Lack of a Retinal Phenotype in a Syne-2/Nesprin-2 Knockout Mouse Model. Cells 2019; 8:cells8101238. [PMID: 31614616 PMCID: PMC6830317 DOI: 10.3390/cells8101238] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Revised: 10/04/2019] [Accepted: 10/10/2019] [Indexed: 01/24/2023] Open
Abstract
Syne-2 (also known as Nesprin-2) is a member of a family of proteins that are found primarily in the outer nuclear membrane, as well as other subcellular compartments. Syne-2 contains a C-terminal KASH transmembrane domain and is part of a protein network that associates the nuclear envelope to the cytoskeleton via the binding to actin filaments. Syne-2 plays a role in nuclear migration, nuclear positioning during retinal development, and in ciliogenesis. In a previous study, we showed a connection between Syne-2 and the multifunctional scaffold protein Pericentrin (Pcnt). The elimination of the interaction of Syne-2 and Pcnt showed defects in nuclear migration and the formation of outer segments during retinal development, as well as disturbances in centrosomal migration at the beginning of ciliogenesis in general. In this study, the Syne-2 KO mouse model Nesprin-2△ABD (Syne-2tm1Ngl, MGI) with special attention to Pcnt and ciliogenesis was analyzed. We show reduced expression of Syne-2 in the retina of the Syne-2 KO mouse but found no significant structural—and only a minor functional—phenotype. For the first time, detailed expression analyses showed an expression of a Syne-2 protein larger than 400 kDa (~750 kDa) in the Syne-2/Nesprin-2 KO mouse. In conclusion, the lack of an overt phenotype in Syne-2/Nesprin-2 KO mice suggests the usage of alternative translational start sites, producing Syne-2 splice variants with an intact Pcnt interaction site. Nevertheless, deletion of the actin-binding site in the Syne-2/Nesprin-2 KO mouse revealed a high variability in scotopic oscillatory potentials assuming a novel function of Syne-2 in synchronizing inner retinal processes.
Collapse
Affiliation(s)
- Nathalie Falk
- Animal Physiology, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91058 Erlangen, Germany.
| | - Anneka Joachimsthaler
- Animal Physiology, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91058 Erlangen, Germany.
- Department of Ophthalmology, University Hospital Erlangen, 91054 Erlangen, Germany.
| | - Kristin Kessler
- Animal Physiology, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91058 Erlangen, Germany.
| | - Uwe Thorsten Lux
- Animal Physiology, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91058 Erlangen, Germany.
| | - Angelika Anna Noegel
- Institute of Biochemistry I; Medical Faculty, University Hospital, University of Cologne, 50931 Cologne, Germany.
| | - Jan Kremers
- Department of Ophthalmology, University Hospital Erlangen, 91054 Erlangen, Germany.
| | | | - Andreas Gießl
- Animal Physiology, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91058 Erlangen, Germany.
- Department of Ophthalmology, University Hospital Erlangen, 91054 Erlangen, Germany.
| | - Nathalie Falk
- Animal Physiology, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91058 Erlangen, Germany
| | - Anneka Joachimsthaler
- Animal Physiology, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91058 Erlangen, Germany
- Department of Ophthalmology, University Hospital Erlangen, 91054 Erlangen, Germany
| | - Kristin Kessler
- Animal Physiology, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91058 Erlangen, Germany
| | - Uwe Thorsten Lux
- Animal Physiology, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91058 Erlangen, Germany
| | - Angelika Anna Noegel
- Institute of Biochemistry I; Medical Faculty, University Hospital, University of Cologne, 50931 Cologne, Germany
| | - Jan Kremers
- Department of Ophthalmology, University Hospital Erlangen, 91054 Erlangen, Germany
| | | | - Andreas Gießl
- Animal Physiology, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91058 Erlangen, Germany.
- Department of Ophthalmology, University Hospital Erlangen, 91054 Erlangen, Germany.
| |
Collapse
|