1
|
Severs LJ, Katta A, Cates LN, Dewees DM, Hoagland RT, Horner PJ, Hofstetter CP, Khaing ZZ. Biomimetic 3D Hydrogels with Aligned Topography for Neural Tissue Engineering. Polymers (Basel) 2024; 16:3556. [PMID: 39771407 PMCID: PMC11678542 DOI: 10.3390/polym16243556] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Revised: 12/11/2024] [Accepted: 12/15/2024] [Indexed: 01/11/2025] Open
Abstract
Spinal cord trauma leads to the destruction of the highly organized cytoarchitecture that carries information along the axis of the spinal column. Currently, there are no clinically accepted strategies that can help regenerate severed axons after spinal cord injury (SCI). Hydrogels are soft biomaterials with high water content that are widely used as scaffolds to interface with the central nervous system (CNS). Here, we examine a simple and reproducible method that results in consistently aligned fibrils within 3D matrices using thermally gelling biomimetic polymers. A collagen type I (Col)-based thermally gelling hydrogel system was used in combination with two other native extracellular matrix proteins: laminin I (LN) and hyaluronic acid (HA). Gelling kinetics for all gel types (Col, Col LN, Col HA) showed that at 37 °C, all three hydrogels formed gels consistently. A method of aspiration and ejection was used to produce Col-based hydrogels containing aligned fibrils. In vitro, embryonic spinal cord neurons survived and produced processes aligned to collagen fibrils. Next, we implanted either non-aligned or aligned hydrogels after a bilateral dorsal hemisection of the thoracic spinal cord at T7/T8. Pan neuronal antibody-positive fibrils were found within all implants; aligned hydrogels supported neurite growth along the parallel direction of the implanted hydrogels. Combined, our in vitro and in vivo data indicate that thermally gelling biomimetic hydrogels can produce aligned matrices through a method of aspiration and ejection, and this presents a novel platform for regenerative therapies for the CNS.
Collapse
Affiliation(s)
- Liza J. Severs
- Department of Physiology and Biophysics, The University of Washington, Seattle, WA 98109, USA;
| | - Anjali Katta
- Department of Neurological Surgery, The University of Washington, Seattle, WA 98109, USA; (A.K.); (L.N.C.); (D.M.D.); (R.T.H.); (C.P.H.)
| | - Lindsay N. Cates
- Department of Neurological Surgery, The University of Washington, Seattle, WA 98109, USA; (A.K.); (L.N.C.); (D.M.D.); (R.T.H.); (C.P.H.)
| | - Dane M. Dewees
- Department of Neurological Surgery, The University of Washington, Seattle, WA 98109, USA; (A.K.); (L.N.C.); (D.M.D.); (R.T.H.); (C.P.H.)
| | - Riana T. Hoagland
- Department of Neurological Surgery, The University of Washington, Seattle, WA 98109, USA; (A.K.); (L.N.C.); (D.M.D.); (R.T.H.); (C.P.H.)
| | - Philip J. Horner
- Department of Neurosurgery, Houston Methodist Research Institute, Houston, TX 98109, USA;
| | - Christoph P. Hofstetter
- Department of Neurological Surgery, The University of Washington, Seattle, WA 98109, USA; (A.K.); (L.N.C.); (D.M.D.); (R.T.H.); (C.P.H.)
| | - Zin Z. Khaing
- Department of Neurological Surgery, The University of Washington, Seattle, WA 98109, USA; (A.K.); (L.N.C.); (D.M.D.); (R.T.H.); (C.P.H.)
| |
Collapse
|
2
|
Sheloukhova L, Watanabe H. Evolution of glial cells: a non-bilaterian perspective. Neural Dev 2024; 19:10. [PMID: 38907299 PMCID: PMC11193209 DOI: 10.1186/s13064-024-00184-4] [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: 09/19/2023] [Accepted: 06/06/2024] [Indexed: 06/23/2024] Open
Abstract
Nervous systems of bilaterian animals generally consist of two cell types: neurons and glial cells. Despite accumulating data about the many important functions glial cells serve in bilaterian nervous systems, the evolutionary origin of this abundant cell type remains unclear. Current hypotheses regarding glial evolution are mostly based on data from model bilaterians. Non-bilaterian animals have been largely overlooked in glial studies and have been subjected only to morphological analysis. Here, we provide a comprehensive overview of conservation of the bilateral gliogenic genetic repertoire of non-bilaterian phyla (Cnidaria, Placozoa, Ctenophora, and Porifera). We overview molecular and functional features of bilaterian glial cell types and discuss their possible evolutionary history. We then examine which glial features are present in non-bilaterians. Of these, cnidarians show the highest degree of gliogenic program conservation and may therefore be crucial to answer questions about glial evolution.
Collapse
Affiliation(s)
- Larisa Sheloukhova
- Evolutionary Neurobiology Unit, Okinawa Institute of Science and Technology, 1919-1 Tancha, Onna-son, Kunigami-gun, Okinawa, 904-0412, Japan
| | - Hiroshi Watanabe
- Evolutionary Neurobiology Unit, Okinawa Institute of Science and Technology, 1919-1 Tancha, Onna-son, Kunigami-gun, Okinawa, 904-0412, Japan.
| |
Collapse
|
3
|
De Bastiani MA, Bellaver B, Carello-Collar G, Zimmermann M, Kunach P, Lima-Filho RA, Forner S, Martini AC, Pascoal TA, Lourenco MV, Rosa-Neto P, Zimmer ER. Cross-species comparative hippocampal transcriptomics in Alzheimer's disease. iScience 2024; 27:108671. [PMID: 38292167 PMCID: PMC10824791 DOI: 10.1016/j.isci.2023.108671] [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: 10/11/2022] [Revised: 07/11/2023] [Accepted: 12/05/2023] [Indexed: 02/01/2024] Open
Abstract
Alzheimer's disease (AD) is a multifactorial pathology, with most cases having a sporadic origin. Recently, knock-in (KI) mouse models, such as the novel humanized amyloid-β (hAβ)-KI, have been developed to better resemble sporadic human AD. METHODS Here, we compared hippocampal publicly available transcriptomic profiles of transgenic (5xFAD and APP/PS1) and KI (hAβ-KI) mouse models with early- (EOAD) and late- (LOAD) onset AD patients. RESULTS The three mouse models presented more Gene Ontology biological processes terms and enriched signaling pathways in common with LOAD than with EOAD individuals. Experimental validation of consistently dysregulated genes revealed five altered in mice (SLC11A1, S100A6, CD14, CD33, and C1QB) and three in humans (S100A6, SLC11A1, and KCNK). Finally, we identified 17 transcription factors potentially acting as master regulators of AD. CONCLUSION Our cross-species analyses revealed that the three mouse models presented a remarkable similarity to LOAD, with the hAβ-KI being the more specific one.
Collapse
Affiliation(s)
- Marco Antônio De Bastiani
- Graduate Program in Biological Sciences: Biochemistry, Department of Biochemistry, Institute of Health Basic Sciences, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, State of Rio Grande do Sul 90035-003, Brazil
| | - Bruna Bellaver
- Department of Psychiatry, School of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Giovanna Carello-Collar
- Graduate Program in Biological Sciences: Biochemistry, Department of Biochemistry, Institute of Health Basic Sciences, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, State of Rio Grande do Sul 90035-003, Brazil
| | - Maria Zimmermann
- Department of Neurology and Neurosurgery, Montréal Neurological Institute, McGill University, Montréal, Québec H3A 1A1, Canada
- Translational Neuroimaging Laboratory, McGill University, Montréal, Québec H4H 1R3, Canada
| | - Peter Kunach
- Department of Neurology and Neurosurgery, Montréal Neurological Institute, McGill University, Montréal, Québec H3A 1A1, Canada
- Translational Neuroimaging Laboratory, McGill University, Montréal, Québec H4H 1R3, Canada
- Douglas Hospital Research Centre, Montreal, Québec H4H 1R3, Canada
| | - Ricardo A.S. Lima-Filho
- Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro, Rio de Janeiro, State of Rio de Janeiro 21941-902, Brazil
| | - Stefania Forner
- Institute for Memory Impairments and Neurological Disorders (UCI MIND), University of California, Irvine, Irvine, CA 92697, USA
| | - Alessandra Cadete Martini
- Department of Pathology & Laboratory Medicine, University of California, Irvine, Irvine, CA 92697, USA
| | - Tharick A. Pascoal
- Department of Psychiatry, School of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
- Department of Neurology, School of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Mychael V. Lourenco
- Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro, Rio de Janeiro, State of Rio de Janeiro 21941-902, Brazil
| | - Pedro Rosa-Neto
- Department of Neurology and Neurosurgery, Montréal Neurological Institute, McGill University, Montréal, Québec H3A 1A1, Canada
- Translational Neuroimaging Laboratory, McGill University, Montréal, Québec H4H 1R3, Canada
- Douglas Hospital Research Centre, Montreal, Québec H4H 1R3, Canada
| | - Eduardo R. Zimmer
- Graduate Program in Biological Sciences: Biochemistry, Department of Biochemistry, Institute of Health Basic Sciences, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, State of Rio Grande do Sul 90035-003, Brazil
- Department of Pharmacology, ICBS, UFRGS, Porto Alegre, State of Rio Grande do Sul 90035-003, Brazil
- Graduate Program in Biological Sciences: Pharmacology and Therapeutics, Department of Pharmacology, ICBS, UFRGS, Porto Alegre, State of Rio Grande do Sul 90035-003, Brazil
- Brain Institute of Rio Grande Do Sul, Pontifical Catholic University of Rio Grande Do Sul, Porto Alegre, State of Rio Grande do Sul 90610-000, Brazil
| |
Collapse
|
4
|
Xie Y, Harwell CC, Garcia ADR. Astrocyte Development in the Rodent. ADVANCES IN NEUROBIOLOGY 2024; 39:51-67. [PMID: 39190071 DOI: 10.1007/978-3-031-64839-7_3] [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
Astrocytes have gained increasing recognition as key elements of a broad array of nervous system functions. These include essential roles in synapse formation and elimination, synaptic modulation, maintenance of the blood-brain barrier, energetic support, and neural repair after injury or disease of the nervous system. Nevertheless, our understanding of mechanisms underlying astrocyte development and maturation remains far behind that of neurons and oligodendrocytes. Early efforts to understand astrocyte development focused primarily on their specification from embryonic progenitors and the molecular mechanisms driving the switch from neuron to glial production. Considerably, less is known about postnatal stages of astrocyte development, the period during which they are predominantly generated and mature. Notably, this period is coincident with synapse formation and the emergence of nascent neural circuits. Thus, a greater understanding of astrocyte development is likely to shed new light on the formation and maturation of synapses and circuits. Here, we highlight key foundational principles of embryonic and postnatal astrocyte development, focusing largely on what is known from rodent studies.
Collapse
Affiliation(s)
- Yajun Xie
- Department of Neurology, University of California, San Francisco, San Francisco, CA, USA
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, San Francisco, CA, USA
| | - Corey C Harwell
- Department of Neurology, University of California, San Francisco, San Francisco, CA, USA
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, San Francisco, CA, USA
| | - A Denise R Garcia
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, PA, USA.
| |
Collapse
|
5
|
Szewczyk LM, Lipiec MA, Liszewska E, Meyza K, Urban-Ciecko J, Kondrakiewicz L, Goncerzewicz A, Rafalko K, Krawczyk TG, Bogaj K, Vainchtein ID, Nakao-Inoue H, Puscian A, Knapska E, Sanders SJ, Jan Nowakowski T, Molofsky AV, Wisniewska MB. Astrocytic β-catenin signaling via TCF7L2 regulates synapse development and social behavior. Mol Psychiatry 2024; 29:57-73. [PMID: 37798419 PMCID: PMC11078762 DOI: 10.1038/s41380-023-02281-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Revised: 09/15/2023] [Accepted: 09/20/2023] [Indexed: 10/07/2023]
Abstract
The Wnt/β-catenin pathway contains multiple high-confidence risk genes that are linked to neurodevelopmental disorders, including autism spectrum disorder. However, its ubiquitous roles across brain cell types and developmental stages have made it challenging to define its impact on neural circuit development and behavior. Here, we show that TCF7L2, which is a key transcriptional effector of the Wnt/β-catenin pathway, plays a cell-autonomous role in postnatal astrocyte maturation and impacts adult social behavior. TCF7L2 was the dominant Wnt effector that was expressed in both mouse and human astrocytes, with a peak during astrocyte maturation. The conditional knockout of Tcf7l2 in postnatal astrocytes led to an enlargement of astrocytes with defective tiling and gap junction coupling. These mice also exhibited an increase in the number of cortical excitatory and inhibitory synapses and a marked increase in social interaction by adulthood. These data reveal an astrocytic role for developmental Wnt/β-catenin signaling in restricting excitatory synapse numbers and regulating adult social behavior.
Collapse
Affiliation(s)
- Lukasz Mateusz Szewczyk
- Department of Psychiatry and Behavioral Sciences/Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA.
- Laboratory of Molecular Neurobiology, Centre of New Technologies, University of Warsaw, Warsaw, Poland.
| | - Marcin Andrzej Lipiec
- Laboratory of Molecular Neurobiology, Centre of New Technologies, University of Warsaw, Warsaw, Poland
- Laboratory of Emotions Neurobiology, BRAINCITY-Center of Excellence for Neural Plasticity and Brain Disorders, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland
| | - Ewa Liszewska
- Laboratory of Molecular and Cellular Neurobiology, International Institute of Molecular and Cell Biology, Warsaw, Poland
| | - Ksenia Meyza
- Laboratory of Emotions Neurobiology, BRAINCITY-Center of Excellence for Neural Plasticity and Brain Disorders, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland
| | - Joanna Urban-Ciecko
- Laboratory of Electrophysiology, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland
| | - Ludwika Kondrakiewicz
- Laboratory of Emotions Neurobiology, BRAINCITY-Center of Excellence for Neural Plasticity and Brain Disorders, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland
| | - Anna Goncerzewicz
- Laboratory of Emotions Neurobiology, BRAINCITY-Center of Excellence for Neural Plasticity and Brain Disorders, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland
| | | | | | - Karolina Bogaj
- Laboratory of Electrophysiology, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland
| | - Ilia Davidovich Vainchtein
- Department of Psychiatry and Behavioral Sciences/Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
- Johnson & Johnson, Neuroscience Therapeutic Area, San Diego, CA, USA
| | - Hiromi Nakao-Inoue
- Department of Psychiatry and Behavioral Sciences/Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
| | - Alicja Puscian
- Laboratory of Emotions Neurobiology, BRAINCITY-Center of Excellence for Neural Plasticity and Brain Disorders, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland
| | - Ewelina Knapska
- Laboratory of Emotions Neurobiology, BRAINCITY-Center of Excellence for Neural Plasticity and Brain Disorders, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland
| | - Stephan J Sanders
- Department of Psychiatry and Behavioral Sciences/Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
- Institute of Developmental and Regenerative Medicine, Department of Paediatrics, University of Oxford, Oxford, OX3 7TY, UK
- New York Genome Center, New York, NY, USA
| | - Tomasz Jan Nowakowski
- Department of Psychiatry and Behavioral Sciences/Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
- Department of Anatomy, University of California, San Francisco, San Francisco, CA, USA
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA, USA
- The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California San Francisco, San Francisco, CA, USA
| | - Anna Victoria Molofsky
- Department of Psychiatry and Behavioral Sciences/Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA.
- Kavli Institute for Fundamental Neuroscience, University of California, San Francisco, San Francisco, CA, USA.
| | - Marta Barbara Wisniewska
- Laboratory of Molecular Neurobiology, Centre of New Technologies, University of Warsaw, Warsaw, Poland.
| |
Collapse
|
6
|
Frith TJR, Briscoe J, Boezio GLM. From signalling to form: the coordination of neural tube patterning. Curr Top Dev Biol 2023; 159:168-231. [PMID: 38729676 DOI: 10.1016/bs.ctdb.2023.11.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/12/2024]
Abstract
The development of the vertebrate spinal cord involves the formation of the neural tube and the generation of multiple distinct cell types. The process starts during gastrulation, combining axial elongation with specification of neural cells and the formation of the neuroepithelium. Tissue movements produce the neural tube which is then exposed to signals that provide patterning information to neural progenitors. The intracellular response to these signals, via a gene regulatory network, governs the spatial and temporal differentiation of progenitors into specific cell types, facilitating the assembly of functional neuronal circuits. The interplay between the gene regulatory network, cell movement, and tissue mechanics generates the conserved neural tube pattern observed across species. In this review we offer an overview of the molecular and cellular processes governing the formation and patterning of the neural tube, highlighting how the remarkable complexity and precision of vertebrate nervous system arises. We argue that a multidisciplinary and multiscale understanding of the neural tube development, paired with the study of species-specific strategies, will be crucial to tackle the open questions.
Collapse
Affiliation(s)
| | - James Briscoe
- The Francis Crick Institute, London, United Kingdom.
| | | |
Collapse
|
7
|
Xie Y, Reid CM, Granados AA, Garcia MT, Dale-Huang F, Hanson SM, Mancia W, Liu J, Adam M, Mosto O, Pisco AO, Alvarez-Buylla A, Harwell CC. Developmental origin and local signals cooperate to determine septal astrocyte identity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.08.561428. [PMID: 37873089 PMCID: PMC10592657 DOI: 10.1101/2023.10.08.561428] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/25/2023]
Abstract
Astrocyte specification during development is influenced by both intrinsic and extrinsic factors, but the precise contribution of each remains poorly understood. Here we show that septal astrocytes from Nkx2.1 and Zic4 expressing progenitor zones are allocated into non-overlapping domains of the medial (MS) and lateral septal nuclei (LS) respectively. Astrocytes in these areas exhibit distinctive molecular and morphological features tailored to the unique cellular and synaptic circuit environment of each nucleus. Using single-nucleus (sn) RNA sequencing, we trace the developmental trajectories of cells in the septum and find that neurons and astrocytes undergo region and developmental stage-specific local cell-cell interactions. We show that expression of the classic morphogens Sonic hedgehog (Shh) and Fibroblast growth factors (Fgfs) by MS and LS neurons respectively, functions to promote the molecular specification of local astrocytes in each region. Finally, using heterotopic cell transplantation, we show that both morphological and molecular specifications of septal astrocytes are highly dependent on the local microenvironment, regardless of developmental origins. Our data highlights the complex interplay between intrinsic and extrinsic factors shaping astrocyte identities and illustrates the importance of the local environment in determining astrocyte functional specialization.
Collapse
Affiliation(s)
- Yajun Xie
- Department of Neurology, University of California, San Francisco, CA
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, San Francisco, CA
| | - Christopher M. Reid
- Department of Neurology, University of California, San Francisco, CA
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, San Francisco, CA
- Department of Neurobiology, Harvard Medical School, Boston, MA
- Ph.D. Program in Neuroscience, Harvard University, Boston, MA
| | | | - Miguel Turrero Garcia
- Department of Neurology, University of California, San Francisco, CA
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, San Francisco, CA
| | - Fiona Dale-Huang
- Department of Neurology, University of California, San Francisco, CA
- Department of Neurological Surgery, University of California, San Francisco, CA
| | - Sarah M. Hanson
- Department of Neurology, University of California, San Francisco, CA
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, San Francisco, CA
| | - Walter Mancia
- Department of Neurology, University of California, San Francisco, CA
- Department of Neurological Surgery, University of California, San Francisco, CA
| | - Jonathan Liu
- Chan Zuckerberg Biohub San Francisco, San Francisco, CA
| | - Manal Adam
- Department of Neurology, University of California, San Francisco, CA
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, San Francisco, CA
| | - Olivia Mosto
- Department of Neurobiology, Harvard Medical School, Boston, MA
| | | | - Arturo Alvarez-Buylla
- Department of Neurology, University of California, San Francisco, CA
- Department of Neurological Surgery, University of California, San Francisco, CA
| | - Corey C. Harwell
- Department of Neurology, University of California, San Francisco, CA
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, San Francisco, CA
- Chan Zuckerberg Biohub San Francisco, San Francisco, CA
- Lead contact
| |
Collapse
|
8
|
Sheloukhova L, Watanabe H. Analysis of cnidarian Gcm suggests a neuronal origin of glial EAAT1 function. Sci Rep 2023; 13:14790. [PMID: 37684386 PMCID: PMC10491807 DOI: 10.1038/s41598-023-42046-9] [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: 05/02/2023] [Accepted: 09/04/2023] [Indexed: 09/10/2023] Open
Abstract
In bilaterian central nervous systems, coordination of neurotransmission by glial cells enables highly sophisticated neural functions. The diversity of transcription factors (TFs) involved in gliogenesis suggests multiple evolutionary origins of various glial cell types of bilaterians. Many of these TFs including the glial cells missing (Gcm) are also present in genomes of Cnidaria, the closest outgroup to Bilateria, but their function remains to be elucidated. In this study, we analyzed the function of Gcm, a multifunctional TF involved in development of glial and non-glial cell types, in the sea anemone, Nematostella vectensis. siRNA-mediated knockdown of Nematostella Gcm altered expression of cell adhesion proteins, glutamate and GABA transporters, ion channels, metabolic enzymes, and zinc finger and Ets-related TFs. NvGcm and mRNAs of downstream genes are expressed in broad neural cell clusters. However, immunostaining of a NvGcm target protein, the glutamate transporter, NvEAAT1, visualized a novel class of cells with flat cell bodies and no clear processes. Together with the finding of unique morphological features of NvEAAT1-functioning cells, these data suggest that extracellular glutamate metabolism, one of major glial functions, is deployed downstream of Gcm in specific neural cell types in Cnidaria.
Collapse
Affiliation(s)
- Larisa Sheloukhova
- Evolutionary Neurobiology Unit, Okinawa Institute of Science and Technology, 1919-1 Tancha, Onna-son, Kunigami-gun, Okinawa, 904-0412, Japan
| | - Hiroshi Watanabe
- Evolutionary Neurobiology Unit, Okinawa Institute of Science and Technology, 1919-1 Tancha, Onna-son, Kunigami-gun, Okinawa, 904-0412, Japan.
| |
Collapse
|
9
|
Markey KM, Saunders JC, Smuts J, von Reyn CR, Garcia ADR. Astrocyte development—More questions than answers. Front Cell Dev Biol 2023; 11:1063843. [PMID: 37051466 PMCID: PMC10083403 DOI: 10.3389/fcell.2023.1063843] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Accepted: 03/14/2023] [Indexed: 03/28/2023] Open
Abstract
The past 15–20 years has seen a remarkable shift in our understanding of astrocyte contributions to central nervous system (CNS) function. Astrocytes have emerged from the shadows of neuroscience and are now recognized as key elements in a broad array of CNS functions. Astrocytes comprise a substantial fraction of cells in the human CNS. Nevertheless, fundamental questions surrounding their basic biology remain poorly understood. While recent studies have revealed a diversity of essential roles in CNS function, from synapse formation and function to blood brain barrier maintenance, fundamental mechanisms of astrocyte development, including their expansion, migration, and maturation, remain to be elucidated. The coincident development of astrocytes and synapses highlights the need to better understand astrocyte development and will facilitate novel strategies for addressing neurodevelopmental and neurological dysfunction. In this review, we provide an overview of the current understanding of astrocyte development, focusing primarily on mammalian astrocytes and highlight outstanding questions that remain to be addressed. We also include an overview of Drosophila glial development, emphasizing astrocyte-like glia given their close anatomical and functional association with synapses. Drosophila offer an array of sophisticated molecular genetic tools and they remain a powerful model for elucidating fundamental cellular and molecular mechanisms governing astrocyte development. Understanding the parallels and distinctions between astrocyte development in Drosophila and vertebrates will enable investigators to leverage the strengths of each model system to gain new insights into astrocyte function.
Collapse
Affiliation(s)
- Kathryn M. Markey
- Department of Biology, Drexel University, Philadelphia, PA, United States
| | | | - Jana Smuts
- Department of Neurobiology and Anatomy, Drexel University, Philadelphia, PA, United States
| | - Catherine R. von Reyn
- Department of Neurobiology and Anatomy, Drexel University, Philadelphia, PA, United States
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, PA, United States
| | - A. Denise R. Garcia
- Department of Biology, Drexel University, Philadelphia, PA, United States
- Department of Neurobiology and Anatomy, Drexel University, Philadelphia, PA, United States
- *Correspondence: A. Denise R. Garcia,
| |
Collapse
|
10
|
Liu Y, Hong W, Gong P, Qi G, Wang X, Kang S, Tang H, Qin S. Specific knockout of Sox2 in astrocytes reduces reactive astrocyte formation and promotes recovery after early postnatal traumatic brain injury in mouse cortex. Glia 2023; 71:602-615. [PMID: 36353976 DOI: 10.1002/glia.24298] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Revised: 10/24/2022] [Accepted: 10/26/2022] [Indexed: 11/11/2022]
Abstract
In response to central nervous system (CNS) injury, astrocytes go through a series of alterations, referred to as reactive astrogliosis, ranging from changes in gene expression and cell hypertrophy to permanent astrocyte borders around stromal cell scars in CNS lesions. The mechanisms underlying injury-induced reactive astrocytes in the adult CNS have been extensively studied. However, little is known about injury-induced reactive astrocytes during early postnatal development. Astrocytes in the mouse cortex are mainly produced through local proliferation during the first 2 weeks after birth. Here we show that Sox2, a transcription factor critical for stem cells and brain development, is expressed in the early postnatal astrocytes and its expression level was increased in reactive astrocytes after traumatic brain injury (TBI) at postnatal day (P) 7 in the cortex. Using a tamoxifen-induced hGFAP-CreERT2; Sox2flox/flox ; Rosa-tdT mouse model, we found that specific knockout of Sox2 in astrocytes greatly inhibited the proliferation of reactive astrocytes, the formation of glia limitans borders and subsequently promoted the tissue recovery after postnatal TBI at P7 in the cortex. In addition, we found that injury-induced glia limitans borders were still formed at P2 in the wild-type mouse cortex, and knockout of Sox2 in astrocytes inhibited the reactivity of both astrocytes and microglia. Together, these findings provide evidence that Sox2 is essential for the reactivity of astrocytes in response to the cortical TBI during the early postnatal period and suggest that Sox2-dependent astrocyte reactivity is a potential target for therapeutic treatment after TBI.
Collapse
Affiliation(s)
- Yitong Liu
- Department of Anatomy, Histology and Embryology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Wentong Hong
- Department of Anatomy, Histology and Embryology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Pifang Gong
- Department of Anatomy, Histology and Embryology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Guibo Qi
- Department of Anatomy, Histology and Embryology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Xiaoxuan Wang
- Department of Anatomy, Histology and Embryology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Siying Kang
- Department of Anatomy, Histology and Embryology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Han Tang
- Department of Anatomy, Histology and Embryology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Song Qin
- Department of Anatomy, Histology and Embryology, School of Basic Medical Sciences, Fudan University, Shanghai, China.,State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China
| |
Collapse
|
11
|
Wiseman JA, Dragunow M, I-H Park T. Cell Type-Specific Nuclei Markers: The Need for Human Brain Research to Go Nuclear. Neuroscientist 2023; 29:41-61. [PMID: 34459315 DOI: 10.1177/10738584211037351] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Identifying and interrogating cell type-specific populations within the heterogeneous milieu of the human brain is paramount to resolving the processes of normal brain homeostasis and the pathogenesis of neurological disorders. While brain cell type-specific markers are well established, most are localized on cellular membranes or within the cytoplasm, with limited literature describing those found in the nucleus. Due to the complex cytoarchitecture of the human brain, immunohistochemical studies require well-defined cell-specific nuclear markers for more precise and efficient quantification of the cellular populations. Furthermore, efficient nuclear markers are required for cell type-specific purification and transcriptomic interrogation of archived human brain tissue through nuclei isolation-based RNA sequencing. To sate the growing demand for robust cell type-specific nuclear markers, we thought it prudent to comprehensively review the current literature to identify and consolidate a novel series of robust cell type-specific nuclear markers that can assist researchers across a range of neuroscientific disciplines. The following review article collates and discusses several key and prospective cell type-specific nuclei markers for each of the major human brain cell types; it then concludes by discussing the potential applications of cell type-specific nuclear workflows and the power of nuclear-based neuroscientific research.
Collapse
Affiliation(s)
- James A Wiseman
- Department of Pharmacology, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand.,Department of Anatomy and Medical Imaging, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Mike Dragunow
- Department of Pharmacology, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand.,Neurosurgical Research Unit, The Centre for Brain Research, University of Auckland, Auckland, New Zealand.,Hugh Green Biobank, The Centre for Brain Research, University of Auckland, Auckland, New Zealand
| | - Thomas I-H Park
- Department of Pharmacology, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand.,Department of Anatomy and Medical Imaging, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| |
Collapse
|
12
|
Su Y, Wang X, Yang Y, Chen L, Xia W, Hoi KK, Li H, Wang Q, Yu G, Chen X, Wang S, Wang Y, Xiao L, Verkhratsky A, Fancy SPJ, Yi C, Niu J. Astrocyte endfoot formation controls the termination of oligodendrocyte precursor cell perivascular migration during development. Neuron 2023; 111:190-201.e8. [PMID: 36384142 PMCID: PMC9922530 DOI: 10.1016/j.neuron.2022.10.032] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Revised: 09/14/2022] [Accepted: 10/22/2022] [Indexed: 11/17/2022]
Abstract
Oligodendrocyte precursor cells (OPCs) undergo an extensive and coordinated migration in the developing CNS, using the pre-formed scaffold of developed blood vessels as their physical substrate for migration. While OPC association with vasculature is critical for dispersal, equally important for permitting differentiation and proper myelination of target axons is their appropriate and timely detachment, but regulation of this process remains unclear. Here we demonstrate a correlation between the developmental formation of astrocytic endfeet on vessels and the termination of OPC perivascular migration. Ex vivo and in vivo live imaging shows that astrocyte endfeet physically displace OPCs from vasculature, and genetic abrogation of endfoot formation hinders both OPC detachment from vessels and subsequent differentiation. Astrocyte-derived semaphorins 3a and 6a act to repel OPCs from blood vessels at the cessation of their perivascular migration and, in so doing, permit subsequent OPC differentiation by insulating them from a maturation inhibitory endothelial niche.
Collapse
Affiliation(s)
- Yixun Su
- Department of Histology and Embryology, Chongqing Key Laboratory of Neurobiology, Brain and Intelligence Research Key Laboratory of Chongqing Education Commission, Third Military Medical University, Chongqing 400038, China; Research Centre, Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen 518107, China
| | - Xiaorui Wang
- Department of Histology and Embryology, Chongqing Key Laboratory of Neurobiology, Brain and Intelligence Research Key Laboratory of Chongqing Education Commission, Third Military Medical University, Chongqing 400038, China
| | - Yujian Yang
- Department of Histology and Embryology, Chongqing Key Laboratory of Neurobiology, Brain and Intelligence Research Key Laboratory of Chongqing Education Commission, Third Military Medical University, Chongqing 400038, China; Department of Ophthalmology, Army Specialty Medical Center, Third Military Medical University, Chongqing 400042, China
| | - Liang Chen
- Department of Orthopedics, Army Specialty Medical Center, Third Military Medical University, Chongqing 400042, China
| | - Wenlong Xia
- Department of Neurology, Department of Pediatrics, Division of Neuroimmunology and Glial Biology, Newborn Brain Research Institute, University of California at San Francisco, San Francisco, CA 94158, USA
| | - Kimberly K Hoi
- Department of Neurology, Department of Pediatrics, Division of Neuroimmunology and Glial Biology, Newborn Brain Research Institute, University of California at San Francisco, San Francisco, CA 94158, USA
| | - Hui Li
- Department of Histology and Embryology, Chongqing Key Laboratory of Neurobiology, Brain and Intelligence Research Key Laboratory of Chongqing Education Commission, Third Military Medical University, Chongqing 400038, China; Research Centre, Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen 518107, China
| | - Qi Wang
- Department of Histology and Embryology, Chongqing Key Laboratory of Neurobiology, Brain and Intelligence Research Key Laboratory of Chongqing Education Commission, Third Military Medical University, Chongqing 400038, China; Research Centre, Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen 518107, China
| | - Guangdan Yu
- Department of Histology and Embryology, Chongqing Key Laboratory of Neurobiology, Brain and Intelligence Research Key Laboratory of Chongqing Education Commission, Third Military Medical University, Chongqing 400038, China
| | - Xiaoying Chen
- Department of Histology and Embryology, Chongqing Key Laboratory of Neurobiology, Brain and Intelligence Research Key Laboratory of Chongqing Education Commission, Third Military Medical University, Chongqing 400038, China
| | - Shouyu Wang
- Department of Histology and Embryology, Chongqing Key Laboratory of Neurobiology, Brain and Intelligence Research Key Laboratory of Chongqing Education Commission, Third Military Medical University, Chongqing 400038, China
| | - Yuxin Wang
- Department of Histology and Embryology, Chongqing Key Laboratory of Neurobiology, Brain and Intelligence Research Key Laboratory of Chongqing Education Commission, Third Military Medical University, Chongqing 400038, China
| | - Lan Xiao
- Department of Histology and Embryology, Chongqing Key Laboratory of Neurobiology, Brain and Intelligence Research Key Laboratory of Chongqing Education Commission, Third Military Medical University, Chongqing 400038, China
| | - Alexei Verkhratsky
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester M139PL, UK; Achucarro Center for Neuroscience, IKERBASQUE, Bilbao 48011, Spain
| | - Stephen P J Fancy
- Department of Neurology, Department of Pediatrics, Division of Neuroimmunology and Glial Biology, Newborn Brain Research Institute, University of California at San Francisco, San Francisco, CA 94158, USA.
| | - Chenju Yi
- Research Centre, Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen 518107, China.
| | - Jianqin Niu
- Department of Histology and Embryology, Chongqing Key Laboratory of Neurobiology, Brain and Intelligence Research Key Laboratory of Chongqing Education Commission, Third Military Medical University, Chongqing 400038, China.
| |
Collapse
|
13
|
Sun J, Song Y, Chen Z, Qiu J, Zhu S, Wu L, Xing L. Heterogeneity and Molecular Markers for CNS Glial Cells Revealed by Single-Cell Transcriptomics. Cell Mol Neurobiol 2022; 42:2629-2642. [PMID: 34704168 PMCID: PMC11421601 DOI: 10.1007/s10571-021-01159-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Accepted: 10/17/2021] [Indexed: 12/11/2022]
Abstract
Glial cells, including astrocytes, oligodendrocytes, and microglia, are the major components in the central nervous system (CNS). Studies have revealed the heterogeneity of each glial cell type and that they each may play distinct roles in physiological processes and/or neurological diseases. Single-cell sequencing (scRNA-seq) technology developed in recent years has extended our understanding of glial cell heterogeneity from the perspective of transcriptome profiling. This review summarizes the marker genes of major glial cells in the CNS and reveals their heterogeneity in different species, CNS regions, developmental stages, and pathological states (Alzheimer's disease and spinal cord injury), expanding our knowledge of glial cell heterogeneity on both molecular and functional levels.
Collapse
Affiliation(s)
- Junjie Sun
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Co-Innovation Center of Neuroregeneration, Nantong University, 19 Qixiu Road, Nantong, 226001, Jiangsu, China
| | - Yixing Song
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Co-Innovation Center of Neuroregeneration, Nantong University, 19 Qixiu Road, Nantong, 226001, Jiangsu, China
| | - Zhiheng Chen
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Co-Innovation Center of Neuroregeneration, Nantong University, 19 Qixiu Road, Nantong, 226001, Jiangsu, China
| | - Jiaying Qiu
- Department of Prenatal Screening and Diagnosis Center, Nantong Maternal and Child Health Hospital affiliated to Nantong University, Nantong, 226001, Jiangsu, China
| | - Shunxing Zhu
- Laboratory Animal Center, Nantong University, Nantong, 226001, China
| | - Liucheng Wu
- Laboratory Animal Center, Nantong University, Nantong, 226001, China.
| | - Lingyan Xing
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Co-Innovation Center of Neuroregeneration, Nantong University, 19 Qixiu Road, Nantong, 226001, Jiangsu, China.
| |
Collapse
|
14
|
Ming Z, Vining B, Bagheri-Fam S, Harley V. SOX9 in organogenesis: shared and unique transcriptional functions. Cell Mol Life Sci 2022; 79:522. [PMID: 36114905 PMCID: PMC9482574 DOI: 10.1007/s00018-022-04543-4] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Revised: 08/13/2022] [Accepted: 08/31/2022] [Indexed: 11/28/2022]
Abstract
The transcription factor SOX9 is essential for the development of multiple organs including bone, testis, heart, lung, pancreas, intestine and nervous system. Mutations in the human SOX9 gene led to campomelic dysplasia, a haploinsufficiency disorder with several skeletal malformations frequently accompanied by 46, XY sex reversal. The mechanisms underlying the diverse SOX9 functions during organ development including its post-translational modifications, the availability of binding partners, and tissue-specific accessibility to target gene chromatin. Here we summarize the expression, activities, and downstream target genes of SOX9 in molecular genetic pathways essential for organ development, maintenance, and function. We also provide an insight into understanding the mechanisms that regulate the versatile roles of SOX9 in different organs.
Collapse
Affiliation(s)
- Zhenhua Ming
- Sex Development Laboratory, Hudson Institute of Medical Research, PO Box 5152, Melbourne, VIC, 3168, Australia
- Department of Molecular and Translational Science, Monash University, Melbourne, VIC, 3800, Australia
| | - Brittany Vining
- Sex Development Laboratory, Hudson Institute of Medical Research, PO Box 5152, Melbourne, VIC, 3168, Australia
- Department of Molecular and Translational Science, Monash University, Melbourne, VIC, 3800, Australia
| | - Stefan Bagheri-Fam
- Sex Development Laboratory, Hudson Institute of Medical Research, PO Box 5152, Melbourne, VIC, 3168, Australia
- Department of Molecular and Translational Science, Monash University, Melbourne, VIC, 3800, Australia
| | - Vincent Harley
- Sex Development Laboratory, Hudson Institute of Medical Research, PO Box 5152, Melbourne, VIC, 3168, Australia.
- Department of Molecular and Translational Science, Monash University, Melbourne, VIC, 3800, Australia.
| |
Collapse
|
15
|
Aleksandrova MA, Sukhinich KK. Astrocytes of the Brain: Retinue Plays the King. Russ J Dev Biol 2022. [DOI: 10.1134/s1062360422040026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
16
|
Duan S, Sawyer TW, Sontz RA, Wieland BA, Diaz AF, Merchant JL. GFAP-directed Inactivation of Men1 Exploits Glial Cell Plasticity in Favor of Neuroendocrine Reprogramming. Cell Mol Gastroenterol Hepatol 2022; 14:1025-1051. [PMID: 35835391 PMCID: PMC9490044 DOI: 10.1016/j.jcmgh.2022.06.009] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Revised: 06/16/2022] [Accepted: 06/28/2022] [Indexed: 01/31/2023]
Abstract
BACKGROUND & AIMS Efforts to characterize the signaling mechanisms that underlie gastroenteropancreatic neoplasms (GEP-NENs) are precluded by a lack of comprehensive models that recapitulate pathogenesis. Investigation into a potential cell-of-origin for gastrin-secreting NENs revealed a non-cell autonomous role for loss of menin in neuroendocrine cell specification, resulting in an induction of gastrin in enteric glia. Here, we investigated the hypothesis that cell autonomous Men1 inactivation in glial fibrillary acidic protein (GFAP)-expressing cells induced neuroendocrine differentiation and tumorigenesis. METHODS Transgenic GFAPΔMen1 mice were generated by conditional GFAP-directed Men1 deletion in GFAP-expressing cells. Cre specificity was confirmed using a tdTomato reporter. GFAPΔMen1 mice were evaluated for GEP-NEN development and neuroendocrine cell hyperplasia. Small interfering RNA-mediated Men1 silencing in a rat enteric glial cell line was performed in parallel. RESULTS GFAPΔMen1 mice developed pancreatic NENs, in addition to pituitary prolactinomas that phenocopied the human MEN1 syndrome. GFAPΔMen1 mice exhibited gastric neuroendocrine hyperplasia that coincided with a significant loss of GFAP expression. Men1 deletion induced loss of glial-restricted progenitor lineage markers and an increase in neuroendocrine genes, suggesting a reprogramming of GFAP+ cells. Deleting Kif3a, a mediator of Hedgehog signaling, in GFAP-expressing cells attenuated neuroendocrine hyperplasia by restricting the neuroendocrine cell fate. Similar results in the pancreas were observed when Sox10 was used to delete Men1. CONCLUSIONS GFAP-directed Men1 inactivation exploits glial cell plasticity in favor of neuroendocrine differentiation.
Collapse
Affiliation(s)
- Suzann Duan
- University of Arizona College of Medicine, Department of Medicine, Division of Gastroenterology, Tucson, Arizona
| | - Travis W. Sawyer
- Wyant College of Optical Sciences, University of Arizona, Tucson, Arizona
| | - Ricky A. Sontz
- University of Arizona College of Medicine, Department of Medicine, Division of Gastroenterology, Tucson, Arizona
| | - Bradley A. Wieland
- University of Arizona College of Medicine, Department of Medicine, Division of Gastroenterology, Tucson, Arizona
| | - Andres F. Diaz
- University of Arizona College of Medicine, Department of Medicine, Division of Gastroenterology, Tucson, Arizona
| | - Juanita L. Merchant
- University of Arizona College of Medicine, Department of Medicine, Division of Gastroenterology, Tucson, Arizona,Correspondence Address correspondence to: Dr Juanita L. Merchant, University of Arizona, 1515 N. Campbell Ave, Tucson, AZ 85724; tel: (520) 626-7897; fax: (520) 626-1291.
| |
Collapse
|
17
|
Lattke M, Guillemot F. Understanding astrocyte differentiation: Clinical relevance, technical challenges, and new opportunities in the omics era. WIREs Mech Dis 2022; 14:e1557. [PMID: 35546493 PMCID: PMC9539907 DOI: 10.1002/wsbm.1557] [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: 05/06/2021] [Revised: 03/23/2022] [Accepted: 03/25/2022] [Indexed: 11/06/2022]
Abstract
Astrocytes are a major type of glial cells that have essential functions in development and homeostasis of the central nervous system (CNS). Immature astrocytes in the developing CNS support neuronal maturation and possess neural-stem-cell-like properties. Mature astrocytes partially lose these functions but gain new functions essential for adult CNS homeostasis. In pathological conditions, astrocytes become "reactive", which disrupts their mature homeostatic functions and reactivates some immature astrocyte-like properties, suggesting a partial reversal of astrocyte maturation. The loss of homeostatic astrocyte functions contributes to the pathogenesis of various neurological conditions, and therefore activating maturation-promoting mechanisms may be a promising therapeutic strategy to restore homeostasis. Manipulating the mechanisms underlying astrocyte maturation might also allow to facilitate CNS regeneration by enhancing developmental functions of adult astrocytes. However, such therapeutic strategies are still some distance away because of our limited understanding of astrocyte differentiation and maturation, due to biological and technical challenges, including the high degree of similarity of astrocytes with neural stem cells and the shortcomings of astrocyte markers. Current advances in systems biology have a huge potential to overcome these challenges. Recent transcriptomic analyses have already revealed new astrocyte markers and new regulators of astrocyte differentiation. However, the epigenomic changes that presumably occur during astrocyte differentiation remain an important, largely unexplored area for future research. Emerging technologies such as CRISPR/Cas9-based functional screens will further improve our understanding of the mechanisms underlying astrocyte differentiation. This may open up new clinical approaches to restore homeostasis in neurological disorders and/or promote CNS regeneration. This article is categorized under: Neurological Diseases > Genetics/Genomics/Epigenetics Neurological Diseases > Stem Cells and Development Neurological Diseases > Molecular and Cellular Physiology.
Collapse
Affiliation(s)
- Michael Lattke
- Neural Stem Cell Biology Laboratory, The Francis Crick Institute, London, UK
| | - Francois Guillemot
- Neural Stem Cell Biology Laboratory, The Francis Crick Institute, London, UK
| |
Collapse
|
18
|
Paredes MF, Mora C, Flores-Ramirez Q, Cebrian-Silla A, Del Dosso A, Larimer P, Chen J, Kang G, Gonzalez Granero S, Garcia E, Chu J, Delgado R, Cotter JA, Tang V, Spatazza J, Obernier K, Ferrer Lozano J, Vento M, Scott J, Studholme C, Nowakowski TJ, Kriegstein AR, Oldham MC, Hasenstaub A, Garcia-Verdugo JM, Alvarez-Buylla A, Huang EJ. Nests of dividing neuroblasts sustain interneuron production for the developing human brain. Science 2022; 375:eabk2346. [PMID: 35084970 DOI: 10.1126/science.abk2346] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The human cortex contains inhibitory interneurons derived from the medial ganglionic eminence (MGE), a germinal zone in the embryonic ventral forebrain. How this germinal zone generates sufficient interneurons for the human brain remains unclear. We found that the human MGE (hMGE) contains nests of proliferative neuroblasts with ultrastructural and transcriptomic features that distinguish them from other progenitors in the hMGE. When dissociated hMGE cells are transplanted into the neonatal mouse brain, they reform into nests containing proliferating neuroblasts that generate young neurons that migrate extensively into the mouse forebrain and mature into different subtypes of functional interneurons. Together, these results indicate that the nest organization and sustained proliferation of neuroblasts in the hMGE provide a mechanism for the extended production of interneurons for the human forebrain.
Collapse
Affiliation(s)
- Mercedes F Paredes
- Department of Neurology, University of California, San Francisco, CA 94143, USA.,Eli and Edythe Broad Institute for Stem Cell Research and Regeneration Medicine, University of California, San Francisco, CA 94143, USA.,Biomedical Sciences Graduate Program, University of California, San Francisco, CA 94143, USA.,Developmental and Stem Cell Graduate Program, University of California, San Francisco, CA 94143, USA
| | - Cristina Mora
- Department of Pathology, University of California, San Francisco, CA 94143, USA
| | | | - Arantxa Cebrian-Silla
- Eli and Edythe Broad Institute for Stem Cell Research and Regeneration Medicine, University of California, San Francisco, CA 94143, USA.,Department of Neurological Surgery, University of California, San Francisco, CA 94143, USA
| | - Ashley Del Dosso
- Department of Pathology, University of California, San Francisco, CA 94143, USA
| | - Phil Larimer
- Department of Neurology, University of California, San Francisco, CA 94143, USA
| | - Jiapei Chen
- Biomedical Sciences Graduate Program, University of California, San Francisco, CA 94143, USA.,Department of Pathology, University of California, San Francisco, CA 94143, USA
| | - Gugene Kang
- Developmental and Stem Cell Graduate Program, University of California, San Francisco, CA 94143, USA.,Department of Neurological Surgery, University of California, San Francisco, CA 94143, USA
| | - Susana Gonzalez Granero
- Laboratorio de Neurobiología Comparada, Instituto Cavanilles de Biodiversidad y Biología Evolutiva, Universitat de València-Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Valencia, Spain
| | - Eric Garcia
- Department of Neurology, University of California, San Francisco, CA 94143, USA
| | - Julia Chu
- Department of Neurology, University of California, San Francisco, CA 94143, USA
| | - Ryan Delgado
- Eli and Edythe Broad Institute for Stem Cell Research and Regeneration Medicine, University of California, San Francisco, CA 94143, USA
| | - Jennifer A Cotter
- Department of Pathology, Children's Hospital Los Angeles, and Keck School of Medicine of University of Southern California, Los Angeles, CA 90027, USA
| | - Vivian Tang
- Department of Pathology, University of California, San Francisco, CA 94143, USA
| | - Julien Spatazza
- Department of Neurological Surgery, University of California, San Francisco, CA 94143, USA
| | - Kirsten Obernier
- Department of Neurological Surgery, University of California, San Francisco, CA 94143, USA
| | - Jaime Ferrer Lozano
- Department of Pathology, Hospital Universitari i Politecnic La Fe, Valencia, Spain
| | - Maximo Vento
- Neonatal Research Group, Health Research Institute La Fe, Valencia, Spain.,Division of Neonatology, University and Polytechnic Hospital La Fe, Valencia, Spain
| | - Julia Scott
- Department of Bioengineering, Santa Clara University, Santa Clara, CA 95053, USA
| | - Colin Studholme
- Biomedical Image Computing Group, Departments of Pediatrics, Bioengineering, and Radiology, University of Washington, Seattle, WA 98195, USA.,Department of Bioengineering, University of Washington, Seattle, WA 98195, USA.,Department of Radiology, University of Washington, Seattle, WA 98195, USA
| | - Tomasz J Nowakowski
- Eli and Edythe Broad Institute for Stem Cell Research and Regeneration Medicine, University of California, San Francisco, CA 94143, USA.,Department of Anatomy and Department of Psychiatry and Behavioral Sciences, University of California, San Francisco, CA 94143, USA
| | - Arnold R Kriegstein
- Department of Neurology, University of California, San Francisco, CA 94143, USA.,Eli and Edythe Broad Institute for Stem Cell Research and Regeneration Medicine, University of California, San Francisco, CA 94143, USA.,Biomedical Sciences Graduate Program, University of California, San Francisco, CA 94143, USA.,Developmental and Stem Cell Graduate Program, University of California, San Francisco, CA 94143, USA
| | - Michael C Oldham
- Developmental and Stem Cell Graduate Program, University of California, San Francisco, CA 94143, USA.,Department of Pathology, University of California, San Francisco, CA 94143, USA.,Department of Neurological Surgery, University of California, San Francisco, CA 94143, USA
| | - Andrea Hasenstaub
- Department of Otolaryngology, University of California, San Francisco, CA 94143, USA
| | - Jose Manuel Garcia-Verdugo
- Laboratorio de Neurobiología Comparada, Instituto Cavanilles de Biodiversidad y Biología Evolutiva, Universitat de València-Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Valencia, Spain
| | - Arturo Alvarez-Buylla
- Eli and Edythe Broad Institute for Stem Cell Research and Regeneration Medicine, University of California, San Francisco, CA 94143, USA.,Biomedical Sciences Graduate Program, University of California, San Francisco, CA 94143, USA.,Developmental and Stem Cell Graduate Program, University of California, San Francisco, CA 94143, USA.,Department of Neurological Surgery, University of California, San Francisco, CA 94143, USA
| | - Eric J Huang
- Eli and Edythe Broad Institute for Stem Cell Research and Regeneration Medicine, University of California, San Francisco, CA 94143, USA.,Biomedical Sciences Graduate Program, University of California, San Francisco, CA 94143, USA.,Developmental and Stem Cell Graduate Program, University of California, San Francisco, CA 94143, USA.,Department of Pathology, University of California, San Francisco, CA 94143, USA
| |
Collapse
|
19
|
Gene Expression of Mouse Hippocampal Stem Cells Grown in a Galactose-Derived Molecular Gel Compared to In Vivo and Neurospheres. Processes (Basel) 2021. [DOI: 10.3390/pr9040716] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Background: N-heptyl-D-galactonamide (GalC7) is a small synthetic carbohydrate derivative that forms a biocompatible supramolecular hydrogel. In this study, the objective was to analyze more in-depth how neural cells differentiate in contact with GalC7. Method: Direct (ex vivo) cells of the fresh hippocampus and culture (In vitro) of the primary cells were investigated. In vitro, investigation performed under three conditions: on culture in neurospheres for 19 days, on culture in GalC7 gel for 7 days, and on culture in both neurospheres and GalC7 gel. Total RNA was isolated with TRIzol from each group, Sox8, Sox9, Sox10, Dcx, and Neurod1 expression levels were measured by qPCR. Result: Sox8 and Sox10, oligodendrocyte markers, and Sox9, an astrocyte marker, were expressed at a much higher level after 7 days of culture in GalC7 hydrogel compared to all other conditions. Dcx, a marker of neurogenesis, and Neurod1, a marker of neuronal differentiation, were expressed at better levels in the GalC7 gel culture compared to the neurosphere. Conclusions: These results show that the GalC7 hydrogel brings different and interesting conditions for inducing the differentiation and maturation of neural progenitor cells compared with polymer-based scaffolds or cell-only conditions. The differences observed open new perspectives in tissue engineering, induction, and transcript analysis.
Collapse
|
20
|
Birck C, Ginolhac A, Pavlou MAS, Michelucci A, Heuschling P, Grandbarbe L. NF-κB and TNF Affect the Astrocytic Differentiation from Neural Stem Cells. Cells 2021; 10:840. [PMID: 33917855 PMCID: PMC8068246 DOI: 10.3390/cells10040840] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 04/02/2021] [Accepted: 04/06/2021] [Indexed: 01/26/2023] Open
Abstract
The NF-κB signaling pathway is crucial during development and inflammatory processes. We have previously shown that NF-κB activation induces dedifferentiation of astrocytes into neural progenitor cells (NPCs). Here, we provide evidence that the NF-κB pathway plays also a fundamental role during the differentiation of NPCs into astrocytes. First, we show that the NF-κB pathway is essential to initiate astrocytic differentiation as its early inhibition induces NPC apoptosis and impedes their differentiation. Second, we demonstrate that persistent NF-κB activation affects NPC-derived astrocyte differentiation. Tumor necrosis factor (TNF)-treated NPCs show NF-κB activation, maintain their multipotential and proliferation properties, display persistent expression of immature markers and inhibit astrocyte markers. Third, we analyze the effect of NF-κB activation on the main known astrocytic differentiation pathways, such as NOTCH and JAK-STAT. Our findings suggest that the NF-κB pathway plays a dual fundamental role during NPC differentiation into astrocytes: it promotes astrocyte specification, but its persistent activation impedes their differentiation.
Collapse
Affiliation(s)
- Cindy Birck
- Department of Life Sciences and Medicine, Faculty of Science, Technology and Communication, University of Luxembourg, L-1511 Luxembourg, Luxembourg; (C.B.); (A.G.); (M.A.S.P.); (P.H.)
| | - Aurélien Ginolhac
- Department of Life Sciences and Medicine, Faculty of Science, Technology and Communication, University of Luxembourg, L-1511 Luxembourg, Luxembourg; (C.B.); (A.G.); (M.A.S.P.); (P.H.)
| | - Maria Angeliki S. Pavlou
- Department of Life Sciences and Medicine, Faculty of Science, Technology and Communication, University of Luxembourg, L-1511 Luxembourg, Luxembourg; (C.B.); (A.G.); (M.A.S.P.); (P.H.)
- NORLUX Neuro-Oncology Laboratory, Department of Oncology, Luxembourg Institute of Health, L-1526 Luxembourg, Luxembourg;
| | - Alessandro Michelucci
- NORLUX Neuro-Oncology Laboratory, Department of Oncology, Luxembourg Institute of Health, L-1526 Luxembourg, Luxembourg;
- Neuro-Immunology Group, Department of Oncology, Luxembourg Institute of Health, L-1526 Luxembourg, Luxembourg
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, L-4362 Esch-sur-Alzette, Luxembourg
| | - Paul Heuschling
- Department of Life Sciences and Medicine, Faculty of Science, Technology and Communication, University of Luxembourg, L-1511 Luxembourg, Luxembourg; (C.B.); (A.G.); (M.A.S.P.); (P.H.)
| | - Luc Grandbarbe
- Department of Life Sciences and Medicine, Faculty of Science, Technology and Communication, University of Luxembourg, L-1511 Luxembourg, Luxembourg; (C.B.); (A.G.); (M.A.S.P.); (P.H.)
| |
Collapse
|
21
|
Abstract
Mouse cortical radial glial cells (RGCs) are primary neural stem cells that give rise to cortical oligodendrocytes, astrocytes, and olfactory bulb (OB) GABAergic interneurons in late embryogenesis. There are fundamental gaps in understanding how these diverse cell subtypes are generated. Here, by combining single-cell RNA-Seq with intersectional lineage analyses, we show that beginning at around E16.5, neocortical RGCs start to generate ASCL1+EGFR+ apical multipotent intermediate progenitors (MIPCs), which then differentiate into basal MIPCs that express ASCL1, EGFR, OLIG2, and MKI67. These basal MIPCs undergo several rounds of divisions to generate most of the cortical oligodendrocytes and astrocytes and a subpopulation of OB interneurons. Finally, single-cell ATAC-Seq supported our model for the genetic logic underlying the specification and differentiation of cortical glial cells and OB interneurons. Taken together, this work reveals the process of cortical radial glial cell lineage progression and the developmental origins of cortical astrocytes and oligodendrocytes.
Collapse
|
22
|
Caramello A, Galichet C, Rizzoti K, Lovell-Badge R. Dentate gyrus development requires a cortical hem-derived astrocytic scaffold. eLife 2021; 10:63904. [PMID: 33393905 PMCID: PMC7806271 DOI: 10.7554/elife.63904] [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: 10/10/2020] [Accepted: 01/01/2021] [Indexed: 01/01/2023] Open
Abstract
During embryonic development, radial glial cells give rise to neurons, then to astrocytes following the gliogenic switch. Timely regulation of the switch, operated by several transcription factors, is fundamental for allowing coordinated interactions between neurons and glia. We deleted the gene for one such factor, SOX9, early during mouse brain development and observed a significantly compromised dentate gyrus (DG). We dissected the origin of the defect, targeting embryonic Sox9 deletion to either the DG neuronal progenitor domain or the adjacent cortical hem (CH). We identified in the latter previously uncharacterized ALDH1L1+ astrocytic progenitors, which form a fimbrial-specific glial scaffold necessary for neuronal progenitor migration toward the developing DG. Our results highlight an early crucial role of SOX9 for DG development through regulation of astroglial potential acquisition in the CH. Moreover, we illustrate how formation of a local network, amidst astrocytic and neuronal progenitors originating from adjacent domains, underlays brain morphogenesis.
Collapse
Affiliation(s)
- Alessia Caramello
- Laboratory of Stem Cell Biology and Developmental Genetics, The Francis Crick Institute, London, United Kingdom
| | - Christophe Galichet
- Laboratory of Stem Cell Biology and Developmental Genetics, The Francis Crick Institute, London, United Kingdom
| | - Karine Rizzoti
- Laboratory of Stem Cell Biology and Developmental Genetics, The Francis Crick Institute, London, United Kingdom
| | - Robin Lovell-Badge
- Laboratory of Stem Cell Biology and Developmental Genetics, The Francis Crick Institute, London, United Kingdom
| |
Collapse
|
23
|
Vaid S, Huttner WB. Transcriptional Regulators and Human-Specific/Primate-Specific Genes in Neocortical Neurogenesis. Int J Mol Sci 2020; 21:ijms21134614. [PMID: 32610533 PMCID: PMC7369782 DOI: 10.3390/ijms21134614] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Revised: 06/09/2020] [Accepted: 06/26/2020] [Indexed: 12/15/2022] Open
Abstract
During development, starting from a pool of pluripotent stem cells, tissue-specific genetic programs help to shape and develop functional organs. To understand the development of an organ and its disorders, it is important to understand the spatio-temporal dynamics of the gene expression profiles that occur during its development. Modifications in existing genes, the de-novo appearance of new genes, or, occasionally, even the loss of genes, can greatly affect the gene expression profile of any given tissue and contribute to the evolution of organs or of parts of organs. The neocortex is evolutionarily the most recent part of the brain, it is unique to mammals, and is the seat of our higher cognitive abilities. Progenitors that give rise to this tissue undergo sequential waves of differentiation to produce the complete sets of neurons and glial cells that make up a functional neocortex. We will review herein our understanding of the transcriptional regulators that control the neural precursor cells (NPCs) during the generation of the most abundant class of neocortical neurons, the glutametergic neurons. In addition, we will discuss the roles of recently-identified human- and primate-specific genes in promoting neurogenesis, leading to neocortical expansion.
Collapse
|
24
|
Abstract
Astrocytes are morphologically complex, ubiquitous cells that are viewed as a homogeneous population tiling the entire central nervous system (CNS). However, this view has been challenged in the last few years with the availability of RNA sequencing, immunohistochemistry, electron microscopy, morphological reconstruction, and imaging data. These studies suggest that astrocytes represent a diverse population of cells and that they display brain area- and disease-specific properties and functions. In this review, we summarize these observations, emphasize areas where clear conclusions can be made, and discuss potential unifying themes. We also identify knowledge gaps that need to be addressed in order to exploit astrocyte diversity as a biological phenomenon of physiological relevance in the CNS. We thus provide a summary and a perspective on astrocyte diversity in the vertebrate CNS.
Collapse
Affiliation(s)
- Baljit S Khakh
- Departments of Physiology and Neurobiology, David Geffen School of Medicine, University of California, Los Angeles, California 90095, USA;
| | - Benjamin Deneen
- Department of Neuroscience and Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, Texas 77030, USA;
| |
Collapse
|
25
|
Marsters CM, Nesan D, Far R, Klenin N, Pittman QJ, Kurrasch DM. Embryonic microglia influence developing hypothalamic glial populations. J Neuroinflammation 2020; 17:146. [PMID: 32375817 PMCID: PMC7201702 DOI: 10.1186/s12974-020-01811-7] [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: 12/16/2019] [Accepted: 04/13/2020] [Indexed: 11/15/2022] Open
Abstract
Background Although historically microglia were thought to be immature in the fetal brain, evidence of purposeful interactions between these immune cells and nearby neural progenitors is becoming established. Here, we examined the influence of embryonic microglia on gliogenesis within the developing tuberal hypothalamus, a region later important for energy balance, reproduction, and thermoregulation. Methods We used immunohistochemistry to quantify the location and numbers of glial cells in the embryonic brain (E13.5–E17.5), as well as a pharmacological approach (i.e., PLX5622) to knock down fetal microglia. We also conducted cytokine and chemokine analyses on embryonic brains in the presence or absence of microglia, and a neurosphere assay to test the effects of the altered cytokines on hypothalamic progenitor behaviors. Results We identified a subpopulation of activated microglia that congregated adjacent to the third ventricle alongside embryonic Olig2+ neural progenitor cells (NPCs) that are destined to give rise to oligodendrocyte and astrocyte populations. In the absence of microglia, we observed an increase in Olig2+ glial progenitor cells that remained at the ventricle by E17.5 and a concomitant decrease of these Olig2+ cells in the mantle zone, indicative of a delay in migration of these precursor cells. A further examination of maturing oligodendrocytes in the hypothalamic grey and white matter area in the absence of microglia revealed migrating oligodendrocyte progenitor cells (OPCs) within the grey matter at E17.5, a time point when OPCs begin to slow their migration. Finally, quantification of cytokine and chemokine signaling in ex vivo E15.5 hypothalamic cultures +/− microglia revealed decreases in the protein levels of several cytokines in the absence of microglia. We assayed the influence of two downregulated cytokines (CCL2 and CXCL10) on neurosphere-forming capacity and lineage commitment of hypothalamic NPCs in culture and showed an increase in NPC proliferation as well as neuronal and oligodendrocyte differentiation. Conclusion These data demonstrate that microglia influence gliogenesis in the developing tuberal hypothalamus.
Collapse
Affiliation(s)
- Candace M Marsters
- Department of Medical Genetics, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada.,Department of Physiology & Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada.,Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada.,Alberta Children's Hospital Research Institute, University of Calgary, Calgary, Alberta, Canada
| | - Dinushan Nesan
- Department of Medical Genetics, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada.,Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada.,Alberta Children's Hospital Research Institute, University of Calgary, Calgary, Alberta, Canada
| | - Rena Far
- Department of Medical Genetics, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada.,Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada.,Alberta Children's Hospital Research Institute, University of Calgary, Calgary, Alberta, Canada
| | - Natalia Klenin
- Department of Medical Genetics, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada.,Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada.,Alberta Children's Hospital Research Institute, University of Calgary, Calgary, Alberta, Canada
| | - Quentin J Pittman
- Department of Physiology & Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada.,Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada.,Alberta Children's Hospital Research Institute, University of Calgary, Calgary, Alberta, Canada
| | - Deborah M Kurrasch
- Department of Medical Genetics, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada. .,Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada. .,Alberta Children's Hospital Research Institute, University of Calgary, Calgary, Alberta, Canada.
| |
Collapse
|
26
|
Abstract
Astrocytes are the most abundant cell type in the central nervous system and have diverse functions in blood–brain barrier maintenance, neural circuitry formation and function, and metabolic regulation. To better understand the diverse roles of astrocytes, we will summarize what is known about astrocyte development and the challenges limiting our understanding of this process. We will also discuss new approaches and technologies advancing the field.
Collapse
Affiliation(s)
- Ekin Su Akdemir
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX, 77030, USA.,Program in Developmental Biology, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Anna Yu-Szu Huang
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX, 77030, USA.,Program in Developmental Biology, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Benjamin Deneen
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX, 77030, USA.,Program in Developmental Biology, Baylor College of Medicine, Houston, TX, 77030, USA.,Department of Neuroscience, Baylor College of Medicine, Houston, TX, 77030, USA.,Department of Neurosurgery, Baylor College of Medicine, Houston, TX, 77030, USA
| |
Collapse
|
27
|
Güven A, Kalebic N, Long KR, Florio M, Vaid S, Brandl H, Stenzel D, Huttner WB. Extracellular matrix-inducing Sox9 promotes both basal progenitor proliferation and gliogenesis in developing neocortex. eLife 2020; 9:49808. [PMID: 32191207 PMCID: PMC7105383 DOI: 10.7554/elife.49808] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2019] [Accepted: 03/18/2020] [Indexed: 12/20/2022] Open
Abstract
Neocortex expansion is largely based on the proliferative capacity of basal progenitors (BPs), which is increased by extracellular matrix (ECM) components via integrin signaling. Here we show that the transcription factor Sox9 drives expression of ECM components and that laminin 211 increases BP proliferation in embryonic mouse neocortex. We show that Sox9 is expressed in human and ferret BPs and is required for BP proliferation in embryonic ferret neocortex. Conditional Sox9 expression in the mouse BP lineage, where it normally is not expressed, increases BP proliferation, reduces Tbr2 levels and induces Olig2 expression, indicative of premature gliogenesis. Conditional Sox9 expression also results in cell-non-autonomous stimulation of BP proliferation followed by increased upper-layer neuron production. Our findings demonstrate that Sox9 exerts concerted effects on transcription, BP proliferation, neuron production, and neurogenic vs. gliogenic BP cell fate, suggesting that Sox9 may have contributed to promote neocortical expansion.
Collapse
Affiliation(s)
- Ayse Güven
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| | - Nereo Kalebic
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany.,Human Technopole, Milan, Italy
| | - Katherine R Long
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| | - Marta Florio
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| | - Samir Vaid
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| | - Holger Brandl
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| | - Denise Stenzel
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| | - Wieland B Huttner
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| |
Collapse
|
28
|
Abstract
Gliomas are heterogeneous tumours derived from glial cells and remain the deadliest form of brain cancer. Although the glioma stem cell sits at the apex of the cellular hierarchy, how it produces the vast cellular constituency associated with frank glioma remains poorly defined. We explore glioma tumorigenesis through the lens of glial development, starting with the neurogenic-gliogenic switch and progressing through oligodendrocyte and astrocyte differentiation. Beginning with the factors that influence normal glial linage progression and diversity, a pattern emerges that has useful parallels in the development of glioma and may ultimately provide targetable pathways for much-needed new therapeutics.
Collapse
|
29
|
Li C, Li M, Li X, Ni W, Xu Y, Yao R, Wei B, Zhang M, Li H, Zhao Y, Liu L, Ullah Y, Jiang Y, Hu S. Whole-Genome Resequencing Reveals Loci Associated With Thoracic Vertebrae Number in Sheep. Front Genet 2019; 10:674. [PMID: 31379930 PMCID: PMC6657399 DOI: 10.3389/fgene.2019.00674] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2019] [Accepted: 06/27/2019] [Indexed: 12/31/2022] Open
Abstract
The number of vertebrae, especially thoracic vertebrae, is an important economic trait that may influence carcass length and meat production in animals. However, the genetic basis of vertebrae number in sheep is still poorly understood. To detect the candidate genes, 400 increased number of thoracic vertebrae (T14L6) and 200 normal (T13L6) Kazakh sheep were collected. We generated and sequenced 60 pools of genomic DNA (each pool prepared by mixing genomic DNA from 10 sheep with the same thoracic traits), with an average depth of coverage of 25.65×. We identified a total of 42,075,402 SNPs and 11 putatively selected genomic regions, including the VRTN gene and the HoxA gene family that regulate vertebral development. The most prominent areas of selective elimination were located in a region of chromosome 7, including VRTN, which regulates spinal development and morphology. Further investigation indicated that the expression level of the VRTN gene during fetal development was significantly higher in sheep with more thoracic vertebrae than in those with a normal number of thoracic vertebrae. A genome-wide comparison between sheep with increased and normal numbers of thoracic vertebrae showed that the VRTN gene is the major selection locus for the number of thoracic vertebrae in sheep and has the potential to be utilized in sheep breeding in the future.
Collapse
Affiliation(s)
- Cunyuan Li
- College of Life Sciences, Shihezi University, Shihezi, China.,College of Animal Science and Technology, Shihezi University, Shihezi, China
| | - Ming Li
- College of Animal Science and Technology, Northwest A&F University, Xianyang, China
| | - Xiaoyue Li
- College of Life Sciences, Shihezi University, Shihezi, China
| | - Wei Ni
- College of Life Sciences, Shihezi University, Shihezi, China
| | - Yueren Xu
- College of Life Sciences, Shihezi University, Shihezi, China
| | - Rui Yao
- College of Life Sciences, Shihezi University, Shihezi, China
| | - Bin Wei
- College of Animal Science and Technology, Northwest A&F University, Xianyang, China
| | - Mengdan Zhang
- College of Life Sciences, Shihezi University, Shihezi, China
| | - Huixiang Li
- College of Life Sciences, Shihezi University, Shihezi, China
| | - Yue Zhao
- College of Animal Science and Technology, Northwest A&F University, Xianyang, China
| | - Li Liu
- College of Life Sciences, Shihezi University, Shihezi, China
| | - Yaseen Ullah
- College of Life Sciences, Shihezi University, Shihezi, China
| | - Yu Jiang
- College of Animal Science and Technology, Northwest A&F University, Xianyang, China
| | - Shengwei Hu
- College of Life Sciences, Shihezi University, Shihezi, China
| |
Collapse
|
30
|
Li J, Khankan RR, Caneda C, Godoy MI, Haney MS, Krawczyk MC, Bassik MC, Sloan SA, Zhang Y. Astrocyte-to-astrocyte contact and a positive feedback loop of growth factor signaling regulate astrocyte maturation. Glia 2019; 67:1571-1597. [PMID: 31033049 PMCID: PMC6557696 DOI: 10.1002/glia.23630] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2018] [Revised: 03/31/2019] [Accepted: 04/05/2019] [Indexed: 01/09/2023]
Abstract
Astrocytes are critical for the development and function of the central nervous system. In developing brains, immature astrocytes undergo morphological, molecular, cellular, and functional changes as they mature. Although the mechanisms that regulate the maturation of other major cell types in the central nervous system such as neurons and oligodendrocytes have been extensively studied, little is known about the cellular and molecular mechanisms that control astrocyte maturation. Here, we identified molecular markers of astrocyte maturation and established an in vitro assay for studying the mechanisms of astrocyte maturation. Maturing astrocytes in vitro exhibit similar molecular changes and represent multiple molecular subtypes of astrocytes found in vivo. Using this system, we found that astrocyte‐to‐astrocyte contact strongly promotes astrocyte maturation. In addition, secreted signals from microglia, oligodendrocyte precursor cells, or endothelial cells affect a small subset of astrocyte genes but do not consistently change astrocyte maturation. To identify molecular mechanisms underlying astrocyte maturation, we treated maturing astrocytes with molecules that affect the function of tumor‐associated genes. We found that a positive feedback loop of heparin‐binding epidermal growth factor‐like growth factor (HBEGF) and epidermal growth factor receptor (EGFR) signaling regulates astrocytes maturation. Furthermore, HBEGF, EGFR, and tumor protein 53 (TP53) affect the expression of genes important for cilium development, the circadian clock, and synapse function. These results revealed cellular and molecular mechanisms underlying astrocytes maturation with implications for the understanding of glioblastoma.
Collapse
Affiliation(s)
- Jiwen Li
- Department of Psychiatry and Biobehavioral Sciences, Semel Institute for Neuroscience and Human Behavior, David Geffen School of Medicine at the University of California, Los Angeles, California
| | - Rana R Khankan
- Department of Psychiatry and Biobehavioral Sciences, Semel Institute for Neuroscience and Human Behavior, David Geffen School of Medicine at the University of California, Los Angeles, California
| | - Christine Caneda
- Department of Psychiatry and Biobehavioral Sciences, Semel Institute for Neuroscience and Human Behavior, David Geffen School of Medicine at the University of California, Los Angeles, California
| | - Marlesa I Godoy
- Department of Psychiatry and Biobehavioral Sciences, Semel Institute for Neuroscience and Human Behavior, David Geffen School of Medicine at the University of California, Los Angeles, California
| | - Michael S Haney
- Department of Genetics, Stanford University School of Medicine, Stanford, California
| | - Mitchell C Krawczyk
- Department of Psychiatry and Biobehavioral Sciences, Semel Institute for Neuroscience and Human Behavior, David Geffen School of Medicine at the University of California, Los Angeles, California
| | - Michael C Bassik
- Department of Genetics, Stanford University School of Medicine, Stanford, California
| | - Steven A Sloan
- Department of Human Genetics, Emory University School of Medicine, Atlanta, Georgia
| | - Ye Zhang
- Department of Psychiatry and Biobehavioral Sciences, Semel Institute for Neuroscience and Human Behavior, David Geffen School of Medicine at the University of California, Los Angeles, California.,Intellectual and Developmental Disabilities Research Center at UCLA, Los Angeles, California.,Brain Research Institute at UCLA, Los Angeles, California.,Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research at UCLA, Los Angeles, California.,Molecular Biology Institute at UCLA, Los Angeles, California
| |
Collapse
|
31
|
Lee MA, Sitko AA, Khalid S, Mason CA. Spatiotemporal distribution of glia in and around the developing mouse optic tract. J Comp Neurol 2019; 527:508-521. [PMID: 29744881 PMCID: PMC6226340 DOI: 10.1002/cne.24462] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2018] [Accepted: 03/21/2018] [Indexed: 12/29/2022]
Abstract
In the developing mouse optic tract, retinal ganglion cell (RGC) axon position is organized by topography and laterality (i.e., eye-specific or ipsi- and contralateral segregation). Our lab previously showed that ipsilaterally projecting RGCs are segregated to the lateral aspect of the developing optic tract and found that ipsilateral axons self-fasciculate to a greater extent than contralaterally projecting RGC axons in vitro. However, the full complement of axon-intrinsic and -extrinsic factors mediating eye-specific segregation in the tract remain poorly understood. Glia, which are known to express several guidance cues in the visual system and regulate the navigation of ipsilateral and contralateral RGC axons at the optic chiasm, are natural candidates for contributing to eye-specific pre-target axon organization. Here, we investigate the spatiotemporal expression patterns of both putative astrocytes (Aldh1l1+ cells) and microglia (Iba1+ cells) in the embryonic and neonatal optic tract. We quantified the localization of ipsilateral RGC axons to the lateral two-thirds of the optic tract and analyzed glia position and distribution relative to eye-specific axon organization. While our results indicate that glial segregation patterns do not strictly align with eye-specific RGC axon segregation in the tract, we identify distinct spatiotemporal organization of both Aldh1l1+ cells and microglia in and around the developing optic tract. These findings inform future research into molecular mechanisms of glial involvement in RGC axon growth and organization in the developing retinogeniculate pathway.
Collapse
Affiliation(s)
- Melissa A. Lee
- Department of Neuroscience, Columbia University, New York, New York, 10027
| | - Austen A. Sitko
- Department of Neurobiology, Harvard Medical School, Boston, Massachusetts, 02115
| | - Sania Khalid
- Department of Pathology and Cell Biology, College of Physicians and Surgeons, Columbia University, New York, New York, 10032
| | - Carol A. Mason
- Department of Neuroscience, Columbia University, New York, New York, 10027
- Department of Pathology and Cell Biology, College of Physicians and Surgeons, Columbia University, New York, New York, 10032
- Department of Ophthalmology, College of Physicians and Surgeons, Columbia University, New York, New York, 10032
| |
Collapse
|
32
|
Miller SJ. Astrocyte Heterogeneity in the Adult Central Nervous System. Front Cell Neurosci 2018; 12:401. [PMID: 30524236 PMCID: PMC6262303 DOI: 10.3389/fncel.2018.00401] [Citation(s) in RCA: 141] [Impact Index Per Article: 20.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2018] [Accepted: 10/18/2018] [Indexed: 11/13/2022] Open
Abstract
Astrocytes are the most abundant cell type in the central nervous system (CNS), providing critical roles in the overall maintenance and homeostasis. Over 100 years ago, Cajal first showed morphological depictions of different astrocyte populations. Surprisingly, to date astrocytes remain classified in two groups based on their morphological and neuroanatomical positioning. However, accumulating evidence over the past few years is showing that astrocytes are highly diverse throughout the CNS. Astrocyte heterogeneity is not surprisingly, as these cells interact with all other cells in the CNS. Like neurons, astrocytes may also have subpopulations that vary in their functionality. In this mini review, we will explore some of the recent evidence in the adult CNS of astrocyte diversity. First, we will review the very little literature on healthy adult astroglia heterogeneity, followed by the identification of different subpopulations in disease states and how this varies between human and mouse. Exploring this new area of neuroscience will hopefully provide researchers with a new perspective on astrocytes and their heterogeneity throughout the CNS.
Collapse
Affiliation(s)
- Sean J Miller
- Laboratory of Tony Wyss-Coray, Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, United States
| |
Collapse
|
33
|
Kelley KW, Nakao-Inoue H, Molofsky AV, Oldham MC. Variation among intact tissue samples reveals the core transcriptional features of human CNS cell classes. Nat Neurosci 2018; 21:1171-1184. [PMID: 30154505 PMCID: PMC6192711 DOI: 10.1038/s41593-018-0216-z] [Citation(s) in RCA: 125] [Impact Index Per Article: 17.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2018] [Accepted: 07/10/2018] [Indexed: 02/08/2023]
Abstract
It is widely assumed that cells must be physically isolated to study their molecular profiles. However, intact tissue samples naturally exhibit variation in cellular composition, which drives covariation of cell-class-specific molecular features. By analyzing transcriptional covariation in 7,221 intact CNS samples from 840 neurotypical individuals, representing billions of cells, we reveal the core transcriptional identities of major CNS cell classes in humans. By modeling intact CNS transcriptomes as a function of variation in cellular composition, we identify cell-class-specific transcriptional differences in Alzheimer's disease, among brain regions, and between species. Among these, we show that PMP2 is expressed by human but not mouse astrocytes and significantly increases mouse astrocyte size upon ectopic expression in vivo, causing them to more closely resemble their human counterparts. Our work is available as an online resource ( http://oldhamlab.ctec.ucsf.edu/ ) and provides a generalizable strategy for determining the core molecular features of cellular identity in intact biological systems.
Collapse
Affiliation(s)
- Kevin W Kelley
- Department of Neurological Surgery, University of California at San Francisco, San Francisco, CA, USA
- The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California at San Francisco, San Francisco, CA, USA
- Weill Institute for Neurosciences, University of California at San Francisco, San Francisco, CA, USA
- Department of Psychiatry, University of California at San Francisco, San Francisco, CA, USA
- Medical Scientist Training Program and Neuroscience Graduate Program, University of California at San Francisco, San Francisco, CA, USA
| | - Hiromi Nakao-Inoue
- The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California at San Francisco, San Francisco, CA, USA
- Weill Institute for Neurosciences, University of California at San Francisco, San Francisco, CA, USA
- Department of Psychiatry, University of California at San Francisco, San Francisco, CA, USA
| | - Anna V Molofsky
- The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California at San Francisco, San Francisco, CA, USA
- Weill Institute for Neurosciences, University of California at San Francisco, San Francisco, CA, USA
- Department of Psychiatry, University of California at San Francisco, San Francisco, CA, USA
| | - Michael C Oldham
- Department of Neurological Surgery, University of California at San Francisco, San Francisco, CA, USA.
- The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California at San Francisco, San Francisco, CA, USA.
- Weill Institute for Neurosciences, University of California at San Francisco, San Francisco, CA, USA.
| |
Collapse
|
34
|
Proliferative cells in the rat developing neocortical grey matter: new insights into gliogenesis. Brain Struct Funct 2018; 223:4053-4066. [PMID: 30132245 DOI: 10.1007/s00429-018-1736-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2018] [Accepted: 08/14/2018] [Indexed: 02/04/2023]
Abstract
The postnatal brain development is characterized by a substantial gain in weight and size, ascribed to increasing neuronal size and branching, and to massive addition of glial cells. This occurs concomitantly to the shrinkage of VZ and SVZ, considered to be the main germinal zones, thus suggesting the existence of other germinative niches. The aim of this study is to characterize the cortical grey matter proliferating cells during postnatal development, providing their stereological quantification and identifying the nature of their cell lineage. We performed double immunolabeling for the proliferation marker Ki67 and three proteins which identify either astrocytes (S100β) or oligodendrocytes (Olig2 and NG2), in addition to a wider panel of markers apt to validate the former markers or to investigate other cell lineages. We found that proliferating cells increase in number during the first postnatal week until P10 and subsequently decreased until P21. Cell lineage characterization revealed that grey matter proliferating cells are prevalently oligodendrocytes and astrocytes along with endothelial and microglial cells, while no neurons have been detected. Our data showed that astrogliogenesis occurs prevalently during the first 10 days of postnatal development, whereas contrary to the expected peak of oligodendrogenesis at the second postnatal week, we found a permanent pool of proliferating oligodendrocytes enduring from birth until P21. These data support the relevance of glial proliferation within the grey matter and could be a point of departure for further investigations of this complex process.
Collapse
|
35
|
Kelley KW, Ben Haim L, Schirmer L, Tyzack GE, Tolman M, Miller JG, Tsai HH, Chang SM, Molofsky AV, Yang Y, Patani R, Lakatos A, Ullian EM, Rowitch DH. Kir4.1-Dependent Astrocyte-Fast Motor Neuron Interactions Are Required for Peak Strength. Neuron 2018; 98:306-319.e7. [PMID: 29606582 PMCID: PMC5919779 DOI: 10.1016/j.neuron.2018.03.010] [Citation(s) in RCA: 88] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2017] [Revised: 11/08/2017] [Accepted: 03/05/2018] [Indexed: 12/11/2022]
Abstract
Diversified neurons are essential for sensorimotor function, but whether astrocytes become specialized to optimize circuit performance remains unclear. Large fast α-motor neurons (FαMNs) of spinal cord innervate fast-twitch muscles that generate peak strength. We report that ventral horn astrocytes express the inward-rectifying K+ channel Kir4.1 (a.k.a. Kcnj10) around MNs in a VGLUT1-dependent manner. Loss of astrocyte-encoded Kir4.1 selectively altered FαMN size and function and led to reduced peak strength. Overexpression of Kir4.1 in astrocytes was sufficient to increase MN size through activation of the PI3K/mTOR/pS6 pathway. Kir4.1 was downregulated cell autonomously in astrocytes derived from amyotrophic lateral sclerosis (ALS) patients with SOD1 mutation. However, astrocyte Kir4.1 was dispensable for FαMN survival even in the mutant SOD1 background. These findings show that astrocyte Kir4.1 is essential for maintenance of peak strength and suggest that Kir4.1 downregulation might uncouple symptoms of muscle weakness from MN cell death in diseases like ALS.
Collapse
Affiliation(s)
- Kevin W Kelley
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA 94143, USA; Departments of Pediatrics and Neurosurgery, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Lucile Ben Haim
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA 94143, USA; Departments of Pediatrics and Neurosurgery, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Lucas Schirmer
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA 94143, USA; Departments of Pediatrics and Neurosurgery, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Giulia E Tyzack
- Department of Molecular Neuroscience, Institute of Neurology, University College London, London WC1N 3BG, UK; The Francis Crick Institute, London NW1 1AT, UK
| | - Michaela Tolman
- Sackler School of Biomedical Sciences, Tufts University, Boston, MA 02111, USA
| | - John G Miller
- Department of Psychiatry, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Hui-Hsin Tsai
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA 94143, USA; Departments of Pediatrics and Neurosurgery, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Sandra M Chang
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA 94143, USA; Departments of Pediatrics and Neurosurgery, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Anna V Molofsky
- Department of Psychiatry, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Yongjie Yang
- Sackler School of Biomedical Sciences, Tufts University, Boston, MA 02111, USA
| | - Rickie Patani
- Department of Molecular Neuroscience, Institute of Neurology, University College London, London WC1N 3BG, UK; The Francis Crick Institute, London NW1 1AT, UK
| | - Andras Lakatos
- John van Geest Centre for Brain Repair and Department of Clinical Neurosciences, University of Cambridge, Cambridge CB20QQ, UK
| | - Erik M Ullian
- Department of Ophthalmology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - David H Rowitch
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA 94143, USA; Departments of Pediatrics and Neurosurgery, University of California, San Francisco, San Francisco, CA 94143, USA; Department of Paediatrics and Wellcome Trust-MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge CB20QQ, UK.
| |
Collapse
|
36
|
Leferink PS, Breeuwsma N, Bugiani M, van der Knaap MS, Heine VM. Affected astrocytes in the spinal cord of the leukodystrophy vanishing white matter. Glia 2018; 66:862-873. [PMID: 29285798 PMCID: PMC5838785 DOI: 10.1002/glia.23289] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2017] [Revised: 12/12/2017] [Accepted: 12/15/2017] [Indexed: 12/24/2022]
Abstract
Leukodystrophies are often devastating diseases, presented with progressive clinical signs as spasticity, ataxia and cognitive decline, and lack proper treatment options. New therapy strategies for leukodystrophies mostly focus on oligodendrocyte replacement to rescue lack of myelin in the brain, even though disease pathology also often involves other glial cells and the spinal cord. In this study we investigated spinal cord pathology in a mouse model for Vanishing White Matter disease (VWM) and show that astrocytes in the white matter are severely affected. Astrocyte pathology starts postnatally in the sensory tracts, followed by changes in the astrocytic populations in the motor tracts. Studies in post-mortem tissue of two VWM patients, a 13-year-old boy and a 6-year-old girl, confirmed astrocyte abnormalities in the spinal cord. For proper development of new treatment options for VWM and, possibly, other leukodystrophies, future studies should investigate spinal cord involvement.
Collapse
Affiliation(s)
- Prisca S. Leferink
- Department of Pediatrics/Child NeurologyAmsterdam Neuroscience, VU University Medical CenterAmsterdamThe Netherlands
| | - Nicole Breeuwsma
- Department of Pediatrics/Child NeurologyAmsterdam Neuroscience, VU University Medical CenterAmsterdamThe Netherlands
| | - Marianna Bugiani
- Department of PathologyVU University Medical Center, Amsterdam NeuroscienceAmsterdamThe Netherlands
| | - Marjo S. van der Knaap
- Department of Pediatrics/Child NeurologyAmsterdam Neuroscience, VU University Medical CenterAmsterdamThe Netherlands
- Department of Functional GenomicsCenter for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, Vrije Universiteit AmsterdamAmsterdamThe Netherlands
| | - Vivi M. Heine
- Department of Pediatrics/Child NeurologyAmsterdam Neuroscience, VU University Medical CenterAmsterdamThe Netherlands
- Department of Complex Trait GeneticsCenter for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, VU Universiteit AmsterdamThe Netherlands
| |
Collapse
|
37
|
Cell Biology of Astrocyte-Synapse Interactions. Neuron 2017; 96:697-708. [PMID: 29096081 DOI: 10.1016/j.neuron.2017.09.056] [Citation(s) in RCA: 692] [Impact Index Per Article: 86.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2017] [Revised: 09/24/2017] [Accepted: 09/28/2017] [Indexed: 12/25/2022]
Abstract
Astrocytes, the most abundant glial cells in the mammalian brain, are critical regulators of brain development and physiology through dynamic and often bidirectional interactions with neuronal synapses. Despite the clear importance of astrocytes for the establishment and maintenance of proper synaptic connectivity, our understanding of their role in brain function is still in its infancy. We propose that this is at least in part due to large gaps in our knowledge of the cell biology of astrocytes and the mechanisms they use to interact with synapses. In this review, we summarize some of the seminal findings that yield important insight into the cellular and molecular basis of astrocyte-neuron communication, focusing on the role of astrocytes in the development and remodeling of synapses. Furthermore, we pose some pressing questions that need to be addressed to advance our mechanistic understanding of the role of astrocytes in regulating synaptic development.
Collapse
|
38
|
Temporal Profiling of Astrocyte Precursors Reveals Parallel Roles for Asef during Development and after Injury. J Neurosci 2017; 36:11904-11917. [PMID: 27881777 DOI: 10.1523/jneurosci.1658-16.2016] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2016] [Revised: 09/12/2016] [Accepted: 09/18/2016] [Indexed: 12/21/2022] Open
Abstract
Lineage development is a stepwise process, governed by stage-specific regulatory factors and associated markers. Astrocytes are one of the principle cell types in the CNS and the stages associated with their development remain very poorly defined. To identify these stages, we performed gene-expression profiling on astrocyte precursor populations in the spinal cord, identifying distinct patterns of gene induction during their development that are strongly correlated with human astrocytes. Validation studies identified a new cohort of astrocyte-associated genes during development and demonstrated their expression in reactive astrocytes in human white matter injury (WMI). Functional studies on one of these genes revealed that mice lacking Asef exhibited impaired astrocyte differentiation during development and repair after WMI, coupled with compromised blood-brain barrier integrity in the adult CNS. These studies have identified distinct stages of astrocyte lineage development associated with human WMI and, together with our functional analysis of Asef, highlight the parallels between astrocyte development and their reactive counterparts associated with injury. SIGNIFICANCE STATEMENT Astrocytes play a central role in CNS function and associated diseases. Yet the mechanisms that control their development remain poorly defined. Using the developing mouse spinal cord as a model system, we identify molecular changes that occur in developing astrocytes. These molecular signatures are strongly correlated with human astrocyte expression profiles and validation in mouse spinal cord identifies a host of new genes associated with the astrocyte lineage. These genes are present in reactive astrocytes in human white matter injury, and functional studies reveal that one of these genes, Asef, contributes to reactive astrocyte responses after injury. These studies identify distinct stages of astrocyte lineage development and highlight the parallels between astrocyte development and their reactive counterparts associated with injury.
Collapse
|
39
|
Yoon H, Walters G, Paulsen AR, Scarisbrick IA. Astrocyte heterogeneity across the brain and spinal cord occurs developmentally, in adulthood and in response to demyelination. PLoS One 2017; 12:e0180697. [PMID: 28700615 PMCID: PMC5507262 DOI: 10.1371/journal.pone.0180697] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2017] [Accepted: 06/20/2017] [Indexed: 01/08/2023] Open
Abstract
Astrocytes have emerged as essential regulators of function and response to injury in the brain and spinal cord, yet very little is known about regional differences that exist. Here we compare the expression of key astroglial markers (glial fibrillary acidic protein (GFAP) and Aldehyde Dehydrogenase-1 Family Member L1 (ALDH1L1)) across these disparate poles of the neuraxis, tracking their expression developmentally and in the context of demyelination. In addition, we document changes in the astrocyte regulatory cytokine interleukin 6 (IL-6), and its signaling partner signal transducer and activator of transcription 3 (STAT3), in vivo and in vitro. Results demonstrate that GFAP expression is higher in the developing and adult spinal cord relative to brain. Comparisons between GFAP and ALDH1L1 expression suggest elevations in spinal cord GFAP during the early postnatal period reflect an accelerated appearance of astrocytes, while elevations in adulthood reflect higher expression by individual astrocytes. Notably, increases in spinal cord compared to whole brain GFAP were paralleled by higher levels of IL-6 and STAT3. Equivalent elevations in GFAP, GFAP/ALDH1L1 ratios, and in IL-6, were observed in primary astrocyte cultures derived from spinal cord compared to cortex. Also, higher levels of GFAP were observed in the spinal cord compared to the brain after focal demyelinating injury. Altogether, these studies point to key differences in astrocyte abundance and the expression of GFAP and IL-6 across the brain and spinal cord that are positioned to influence regional specialization developmentally and responses occurring in the context of injury and disease.
Collapse
Affiliation(s)
- Hyesook Yoon
- Department of Physical Medicine and Rehabilitation, Rehabilitation Medicine Research Center, Mayo Clinic, Rochester, Minnesota, United States of America
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Grant Walters
- Department of Physical Medicine and Rehabilitation, Rehabilitation Medicine Research Center, Mayo Clinic, Rochester, Minnesota, United States of America
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Alex R. Paulsen
- Department of Physical Medicine and Rehabilitation, Rehabilitation Medicine Research Center, Mayo Clinic, Rochester, Minnesota, United States of America
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Isobel A. Scarisbrick
- Department of Physical Medicine and Rehabilitation, Rehabilitation Medicine Research Center, Mayo Clinic, Rochester, Minnesota, United States of America
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota, United States of America
- Neurobiology of Disease Program, Mayo Clinic, Rochester, Minnesota, United States of America
- * E-mail:
| |
Collapse
|
40
|
Seidel K, Marangoni P, Tang C, Houshmand B, Du W, Maas RL, Murray S, Oldham MC, Klein OD. Resolving stem and progenitor cells in the adult mouse incisor through gene co-expression analysis. eLife 2017; 6:e24712. [PMID: 28475038 PMCID: PMC5419740 DOI: 10.7554/elife.24712] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2016] [Accepted: 04/07/2017] [Indexed: 12/12/2022] Open
Abstract
Investigations into stem cell-fueled renewal of an organ benefit from an inventory of cell type-specific markers and a deep understanding of the cellular diversity within stem cell niches. Using the adult mouse incisor as a model for a continuously renewing organ, we performed an unbiased analysis of gene co-expression relationships to identify modules of co-expressed genes that represent differentiated cells, transit-amplifying cells, and residents of stem cell niches. Through in vivo lineage tracing, we demonstrated the power of this approach by showing that co-expression module members Lrig1 and Igfbp5 define populations of incisor epithelial and mesenchymal stem cells. We further discovered that two adjacent mesenchymal tissues, the periodontium and dental pulp, are maintained by distinct pools of stem cells. These findings reveal novel mechanisms of incisor renewal and illustrate how gene co-expression analysis of intact biological systems can provide insights into the transcriptional basis of cellular identity.
Collapse
Affiliation(s)
- Kerstin Seidel
- Department of Orofacial Sciences and Program in Craniofacial BiologyUniversity of California, San FranciscoSan FranciscoUnited States
| | - Pauline Marangoni
- Department of Orofacial Sciences and Program in Craniofacial BiologyUniversity of California, San FranciscoSan FranciscoUnited States
| | - Cynthia Tang
- Department of Orofacial Sciences and Program in Craniofacial BiologyUniversity of California, San FranciscoSan FranciscoUnited States
| | - Bahar Houshmand
- Department of Orofacial Sciences and Program in Craniofacial BiologyUniversity of California, San FranciscoSan FranciscoUnited States
| | - Wen Du
- Department of Orofacial Sciences and Program in Craniofacial BiologyUniversity of California, San FranciscoSan FranciscoUnited States
| | - Richard L Maas
- Division of Genetics, Department of MedicineBrigham and Women’s Hospital, Harvard Medical SchoolBostonUnited States
| | | | - Michael C Oldham
- Department of Neurological SurgeryUniversity of California, San FranciscoSan FranciscoUnited States
- The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell ResearchUniversity of California, San FranciscoSan FranciscoUnited States
| | - Ophir D Klein
- Department of Orofacial Sciences and Program in Craniofacial BiologyUniversity of California, San FranciscoSan FranciscoUnited States
- The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell ResearchUniversity of California, San FranciscoSan FranciscoUnited States
- Department of Pediatrics and Institute for Human GeneticsUniversity of California, San FranciscoSan FranciscoUnited States
| |
Collapse
|
41
|
Reiprich S, Cantone M, Weider M, Baroti T, Wittstatt J, Schmitt C, Küspert M, Vera J, Wegner M. Transcription factor Sox10 regulates oligodendroglial Sox9 levels via microRNAs. Glia 2017; 65:1089-1102. [PMID: 28370559 DOI: 10.1002/glia.23146] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2017] [Revised: 03/15/2017] [Accepted: 03/16/2017] [Indexed: 01/12/2023]
Abstract
During development of myelin-forming oligodendrocytes in the central nervous system the two closely related transcription factors Sox9 and Sox10 play essential roles that are partly shared and partly unique. Whereas Sox9 primarily functions during oligodendroglial specification, Sox10 is uniquely required to induce terminal differentiation and myelination. During this process, Sox10 protein levels rise substantially. As this coincides with a reciprocal decrease in Sox9, we postulated that Sox10 influences Sox9 amounts in differentiating oligodendrocytes. Here we show that Sox9 levels are indeed inversely coupled to Sox10 levels such that Sox10 deletion in oligodendroglial cells evokes a reciprocal increase in Sox9. We furthermore provide evidence that this coupling involves upregulation of microRNAs miR335 and miR338 as direct transcriptional targets of Sox10. The two microRNAs in turn recognize the 3'-UTR of Sox9 mRNA and may thereby reduce Sox9 protein levels posttranscriptionally in oligodendroglial cells. Such a mechanism may enable oligodendroglial cells to adapt the ratio of both related Sox proteins in a manner required for successful lineage progression and differentiation. Mathematical modeling furthermore shows that the identified regulatory circuit has the potential to convert a transient stimulus into an irreversible switch of cellular properties and may thus contribute to terminal differentiation of oligodendrocytes.
Collapse
Affiliation(s)
- Simone Reiprich
- Institut für Biochemie, Emil-Fischer-Zentrum, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Martina Cantone
- Department of Dermatology, Laboratory of Systems Tumor Immunology, Erlangen University Hospital, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Matthias Weider
- Institut für Biochemie, Emil-Fischer-Zentrum, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Tina Baroti
- Institut für Biochemie, Emil-Fischer-Zentrum, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Jan Wittstatt
- Institut für Biochemie, Emil-Fischer-Zentrum, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Christian Schmitt
- Institut für Biochemie, Emil-Fischer-Zentrum, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Melanie Küspert
- Institut für Biochemie, Emil-Fischer-Zentrum, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Julio Vera
- Department of Dermatology, Laboratory of Systems Tumor Immunology, Erlangen University Hospital, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Michael Wegner
- Institut für Biochemie, Emil-Fischer-Zentrum, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| |
Collapse
|
42
|
SOX9 Is an Astrocyte-Specific Nuclear Marker in the Adult Brain Outside the Neurogenic Regions. J Neurosci 2017; 37:4493-4507. [PMID: 28336567 DOI: 10.1523/jneurosci.3199-16.2017] [Citation(s) in RCA: 242] [Impact Index Per Article: 30.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2016] [Revised: 01/25/2017] [Accepted: 02/19/2017] [Indexed: 01/23/2023] Open
Abstract
Astrocytes have in recent years become the focus of intense experimental interest, yet markers for their definitive identification remain both scarce and imperfect. Astrocytes may be recognized as such by their expression of glial fibrillary acidic protein, glutamine synthetase, glutamate transporter 1 (GLT1), aquaporin-4, aldehyde dehydrogenase 1 family member L1, and other proteins. However, these proteins may all be regulated both developmentally and functionally, restricting their utility. To identify a nuclear marker pathognomonic of astrocytic phenotype, we assessed differential RNA expression by FACS-purified adult astrocytes and, on that basis, evaluated the expression of the transcription factor SOX9 in both mouse and human brain. We found that SOX9 is almost exclusively expressed by astrocytes in the adult brain except for ependymal cells and in the neurogenic regions, where SOX9 is also expressed by neural progenitor cells. Transcriptome comparisons of SOX9+ cells with GLT1+ cells showed that the two populations of cells exhibit largely overlapping gene expression. Expression of SOX9 did not decrease during aging and was instead upregulated by reactive astrocytes in a number of settings, including a murine model of amyotrophic lateral sclerosis (SOD1G93A), middle cerebral artery occlusion, and multiple mini-strokes. We quantified the relative number of astrocytes using the isotropic fractionator technique in combination with SOX9 immunolabeling. The analysis showed that SOX9+ astrocytes constitute ∼10-20% of the total cell number in most CNS regions, a smaller fraction of total cell number than previously estimated in the normal adult brain.SIGNIFICANCE STATEMENT Astrocytes are traditionally identified immunohistochemically by antibodies that target cell-specific antigens in the cytosol or plasma membrane. We show here that SOX9 is an astrocyte-specific nuclear marker in all major areas of the CNS outside of the neurogenic regions. Based on SOX9 immunolabeling, we document that astrocytes constitute a smaller fraction of total cell number than previously estimated in the normal adult mouse brain.
Collapse
|
43
|
Alexovič Matiašová A, Ševc J, Tomori Z, Gombalová Z, Gedrová Š, Daxnerová Z. Quantitative analyses of cellularity and proliferative activity reveals the dynamics of the central canal lining during postnatal development of the rat. J Comp Neurol 2017; 525:693-707. [PMID: 27503700 DOI: 10.1002/cne.24091] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2015] [Revised: 07/12/2016] [Accepted: 08/03/2016] [Indexed: 11/07/2022]
Abstract
According to previous opinion, the derivation of neurons and glia from the central canal (CC) lining of the spinal cord in rodents should occur in the embryonic period. Reports of the mitotic activity observed in the lining during postnatal development have often been contradictory, and proliferation was ascribed to the generation of ependymocytes, which are necessary for the elongation of CC walls. Our study quantifies the intensity of proliferation and determines the cellularity of the CC lining in reference to lumbar spinal segment L4 during the postnatal development of rats. The presence of dividing cells peaks in the CC lining on postnatal day 8 (P8), with division occurring in 19.2% ± 3.2% of cells. In adult rats, 3.6% ± 0.9% of cells still proliferate, whereas, in mice, 10.3% ± 2.3% of cells at P8 and only 0.6% ± 0.2% of cells in the CC lining in adulthood are proliferating. In the rat, the length of the cell cycle increases from 100.3 ± 35.7 hours at P1 to 401.4 ± 80.6 hours at P43, with a sudden extension between P15 and P22. Despite the intensive proliferation, the total cellularity of the CC lining at the L4 spinal segment significantly descended in from P8 to P15. According to our calculations, the estimated cellularity was significantly higher compared with the measured cellularity of the CC lining at P15. Our results indicate that CC lining serves as a source of cells beyond ependymal cells during the first postnatal weeks of the rat. J. Comp. Neurol. 525:693-707, 2017. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Anna Alexovič Matiašová
- Institute of Biology and Ecology, Faculty of Science, P.J. Šafárik University in Košice, 04001, Košice, Slovak Republic
| | - Juraj Ševc
- Institute of Biology and Ecology, Faculty of Science, P.J. Šafárik University in Košice, 04001, Košice, Slovak Republic
| | - Zoltán Tomori
- Institute of Experimental Physics, Slovak Academy of Sciences, 04001, Košice, Slovak Republic
| | - Zuzana Gombalová
- Institute of Biology and Ecology, Faculty of Science, P.J. Šafárik University in Košice, 04001, Košice, Slovak Republic
| | - Štefánia Gedrová
- Institute of Biology and Ecology, Faculty of Science, P.J. Šafárik University in Košice, 04001, Košice, Slovak Republic
| | - Zuzana Daxnerová
- Institute of Biology and Ecology, Faculty of Science, P.J. Šafárik University in Košice, 04001, Košice, Slovak Republic
| |
Collapse
|
44
|
Ependymal cell contribution to scar formation after spinal cord injury is minimal, local and dependent on direct ependymal injury. Sci Rep 2017; 7:41122. [PMID: 28117356 PMCID: PMC5259707 DOI: 10.1038/srep41122] [Citation(s) in RCA: 83] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2016] [Accepted: 12/14/2016] [Indexed: 01/30/2023] Open
Abstract
Ependyma have been proposed as adult neural stem cells that provide the majority of newly proliferated scar-forming astrocytes that protect tissue and function after spinal cord injury (SCI). This proposal was based on small, midline stab SCI. Here, we tested the generality of this proposal by using a genetic knock-in cell fate mapping strategy in different murine SCI models. After large crush injuries across the entire spinal cord, ependyma-derived progeny remained local, did not migrate and contributed few cells of any kind and less than 2%, if any, of the total newly proliferated and molecularly confirmed scar-forming astrocytes. Stab injuries that were near to but did not directly damage ependyma, contained no ependyma-derived cells. Our findings show that ependymal contribution of progeny after SCI is minimal, local and dependent on direct ependymal injury, indicating that ependyma are not a major source of endogenous neural stem cells or neuroprotective astrocytes after SCI.
Collapse
|
45
|
Srinivasan R, Lu TY, Chai H, Xu J, Huang BS, Golshani P, Coppola G, Khakh BS. New Transgenic Mouse Lines for Selectively Targeting Astrocytes and Studying Calcium Signals in Astrocyte Processes In Situ and In Vivo. Neuron 2016; 92:1181-1195. [PMID: 27939582 PMCID: PMC5403514 DOI: 10.1016/j.neuron.2016.11.030] [Citation(s) in RCA: 302] [Impact Index Per Article: 33.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2016] [Revised: 10/14/2016] [Accepted: 11/15/2016] [Indexed: 11/23/2022]
Abstract
Astrocytes exist throughout the nervous system and are proposed to affect neural circuits and behavior. However, studying astrocytes has proven difficult because of the lack of tools permitting astrocyte-selective genetic manipulations. Here, we report the generation of Aldh1l1-Cre/ERT2 transgenic mice to selectively target astrocytes in vivo. We characterized Aldh1l1-Cre/ERT2 mice using imaging, immunohistochemistry, AAV-FLEX-GFP microinjections, and crosses to RiboTag, Ai95, and new Cre-dependent membrane-tethered Lck-GCaMP6f knockin mice that we also generated. Two to three weeks after tamoxifen induction, Aldh1l1-Cre/ERT2 selectively targeted essentially all adult (P80) brain astrocytes with no detectable neuronal contamination, resulting in expression of cytosolic and Lck-GCaMP6f, and permitting subcellular astrocyte calcium imaging during startle responses in vivo. Crosses with RiboTag mice allowed sequencing of actively translated mRNAs and determination of the adult cortical astrocyte transcriptome. Thus, we provide well-characterized, easy-to-use resources with which to selectively study astrocytes in situ and in vivo in multiple experimental scenarios.
Collapse
Affiliation(s)
- Rahul Srinivasan
- Department of Physiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095-1751, USA
| | - Tsai-Yi Lu
- Department of Physiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095-1751, USA
| | - Hua Chai
- Department of Physiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095-1751, USA
| | - Ji Xu
- Department of Physiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095-1751, USA
| | - Ben S Huang
- Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095-1751, USA
| | - Peyman Golshani
- Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095-1751, USA; Department of Psychiatry and Biobehavioral Sciences, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095-1751, USA; Integrative Center for Learning and Memory, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095-1751, USA; West Los Angeles VA Medical Center, Los Angeles, CA 90073, USA
| | - Giovanni Coppola
- Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095-1751, USA; Department of Psychiatry and Biobehavioral Sciences, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095-1751, USA; Center for Neurobehavioral Genetics, Semel Institute for Neuroscience and Human Behavior, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095-1751, USA
| | - Baljit S Khakh
- Department of Physiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095-1751, USA; Department of Neurobiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095-1751, USA.
| |
Collapse
|
46
|
Molecular and cellular basis for the unique functioning of Nrf1, an indispensable transcription factor for maintaining cell homoeostasis and organ integrity. Biochem J 2016; 473:961-1000. [PMID: 27060105 DOI: 10.1042/bj20151182] [Citation(s) in RCA: 101] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2015] [Accepted: 01/26/2016] [Indexed: 12/30/2022]
Abstract
The consensuscis-regulatory AP-1 (activator protein-1)-like AREs (antioxidant-response elements) and/or EpREs (electrophile-response elements) allow for differential recruitment of Nrf1 [NF-E2 (nuclear factor-erythroid 2)-related factor 1], Nrf2 and Nrf3, together with each of their heterodimeric partners (e.g. sMaf, c-Jun, JunD or c-Fos), to regulate different sets of cognate genes. Among them, NF-E2 p45 and Nrf3 are subject to tissue-specific expression in haemopoietic and placental cell lineages respectively. By contrast, Nrf1 and Nrf2 are two important transcription factors expressed ubiquitously in various vertebrate tissues and hence may elicit putative combinational or competitive functions. Nevertheless, they have de facto distinct biological activities because knockout of their genes in mice leads to distinguishable phenotypes. Of note, Nrf2 is dispensable during development and growth, albeit it is accepted as a master regulator of antioxidant, detoxification and cytoprotective genes against cellular stress. Relative to the water-soluble Nrf2, less attention has hitherto been drawn to the membrane-bound Nrf1, even though it has been shown to be indispensable for embryonic development and organ integrity. The biological discrepancy between Nrf1 and Nrf2 is determined by differences in both their primary structures and topovectorial subcellular locations, in which they are subjected to distinct post-translational processing so as to mediate differential expression of ARE-driven cytoprotective genes. In the present review, we focus on the molecular and cellular basis for Nrf1 and its isoforms, which together exert its essential functions for maintaining cellular homoeostasis, normal organ development and growth during life processes. Conversely, dysfunction of Nrf1 results in spontaneous development of non-alcoholic steatohepatitis, hepatoma, diabetes and neurodegenerative diseases in animal models.
Collapse
|
47
|
Weider M, Wegner M. SoxE factors: Transcriptional regulators of neural differentiation and nervous system development. Semin Cell Dev Biol 2016; 63:35-42. [PMID: 27552919 DOI: 10.1016/j.semcdb.2016.08.013] [Citation(s) in RCA: 85] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2016] [Revised: 08/16/2016] [Accepted: 08/18/2016] [Indexed: 12/20/2022]
Abstract
Sox8, Sox9 and Sox10 represent the three vertebrate members of the SoxE subclass of high-mobility-group domain containing Sox transcription factors. They play important roles in the peripheral and central nervous systems as regulators of stemness, specification, survival, lineage progression, glial differentiation and homeostasis. Functions are frequently overlapping, but sometimes antagonistic. SoxE proteins dynamically interact with transcriptional regulators, chromatin changing complexes and components of the transcriptional machinery. By establishing regulatory circuits with other transcription factors and microRNAs, SoxE proteins perform divergent functions in several cell lineages of the vertebrate nervous system, and at different developmental stages in the same cell lineage. The underlying molecular mechanisms are the topic of this review.
Collapse
Affiliation(s)
- Matthias Weider
- Institut für Biochemie, Emil-Fischer-Zentrum, Friedrich-Alexander-Universität Erlangen-Nürnberg, D-91054 Erlangen, Germany
| | - Michael Wegner
- Institut für Biochemie, Emil-Fischer-Zentrum, Friedrich-Alexander-Universität Erlangen-Nürnberg, D-91054 Erlangen, Germany.
| |
Collapse
|
48
|
Molofsky AB, Savage AK, Locksley RM. Interleukin-33 in Tissue Homeostasis, Injury, and Inflammation. Immunity 2015; 42:1005-19. [PMID: 26084021 DOI: 10.1016/j.immuni.2015.06.006] [Citation(s) in RCA: 491] [Impact Index Per Article: 49.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2015] [Indexed: 12/12/2022]
Abstract
Interleukin-33 (IL-33) is a nuclear-associated cytokine of the IL-1 family originally described as a potent inducer of allergic type 2 immunity. IL-33 signals via the receptor ST2, which is highly expressed on group 2 innate lymphoid cells (ILC2s) and T helper 2 (Th2) cells, thus underpinning its association with helminth infection and allergic pathology. Recent studies have revealed ST2 expression on subsets of regulatory T cells, and for a role for IL-33 in tissue homeostasis and repair that suggests previously unrecognized interactions within these cellular networks. IL-33 can participate in pathologic fibrotic reactions, or, in the setting of microbial invasion, can cooperate with inflammatory cytokines to promote responses by cytotoxic NK cells, Th1 cells, and CD8(+) T cells. Here, we highlight the regulation and function of IL-33 and ST2 and review their roles in homeostasis, damage, and inflammation, suggesting a conceptual framework for future studies.
Collapse
Affiliation(s)
- Ari B Molofsky
- Department of Microbiology & Immunology, University of California, San Francisco, 94143-0795, USA; Department of Laboratory Medicine, University of California, San Francisco, 94143-0795, USA
| | - Adam K Savage
- Howard Hughes Medical Institute, University of California, San Francisco, 94143-0795, USA; Department of Microbiology & Immunology, University of California, San Francisco, 94143-0795, USA
| | - Richard M Locksley
- Howard Hughes Medical Institute, University of California, San Francisco, 94143-0795, USA; Department of Medicine, University of California, San Francisco, 94143-0795, USA; Department of Microbiology & Immunology, University of California, San Francisco, 94143-0795, USA.
| |
Collapse
|
49
|
Nagao M, Lanjakornsiripan D, Itoh Y, Kishi Y, Ogata T, Gotoh Y. High mobility group nucleosome-binding family proteins promote astrocyte differentiation of neural precursor cells. Stem Cells 2015; 32:2983-97. [PMID: 25069414 DOI: 10.1002/stem.1787] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2014] [Revised: 06/17/2014] [Accepted: 06/23/2014] [Indexed: 11/07/2022]
Abstract
Astrocytes are the most abundant cell type in the mammalian brain and are important for the functions of the central nervous system. Although previous studies have shown that the STAT signaling pathway or its regulators promote the generation of astrocytes from multipotent neural precursor cells (NPCs) in the developing mammalian brain, the molecular mechanisms that regulate the astrocytic fate decision have still remained largely unclear. Here, we show that the high mobility group nucleosome-binding (HMGN) family proteins, HMGN1, 2, and 3, promote astrocyte differentiation of NPCs during brain development. HMGN proteins were expressed in NPCs, Sox9(+) glial progenitors, and GFAP(+) astrocytes in perinatal and adult brains. Forced expression of either HMGN1, 2, or 3 in NPCs in cultures or in the late embryonic neocortex increased the generation of astrocytes at the expense of neurons. Conversely, knockdown of either HMGN1, 2, or 3 in NPCs suppressed astrocyte differentiation and promoted neuronal differentiation. Importantly, overexpression of HMGN proteins did not induce the phosphorylation of STAT3 or activate STAT reporter genes. In addition, HMGN family proteins did not enhance DNA demethylation and acetylation of histone H3 around the STAT-binding site of the gfap promoter. Moreover, knockdown of HMGN family proteins significantly reduced astrocyte differentiation induced by gliogenic signal ciliary neurotrophic factor, which activates the JAK-STAT pathway. Therefore, we propose that HMGN family proteins are novel chromatin regulatory factors that control astrocyte fate decision/differentiation in parallel with or downstream of the JAK-STAT pathway through modulation of the responsiveness to gliogenic signals.
Collapse
Affiliation(s)
- Motoshi Nagao
- Department of Rehabilitation for the Movement Functions, Research Institute, National Rehabilitation Center for Persons with Disabilities, Saitama, Japan
| | | | | | | | | | | |
Collapse
|
50
|
Zhao C, Ma D, Zawadzka M, Fancy SPJ, Elis-Williams L, Bouvier G, Stockley JH, de Castro GM, Wang B, Jacobs S, Casaccia P, Franklin RJM. Sox2 Sustains Recruitment of Oligodendrocyte Progenitor Cells following CNS Demyelination and Primes Them for Differentiation during Remyelination. J Neurosci 2015; 35:11482-99. [PMID: 26290228 PMCID: PMC6605237 DOI: 10.1523/jneurosci.3655-14.2015] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2014] [Revised: 05/27/2015] [Accepted: 06/21/2015] [Indexed: 12/16/2022] Open
Abstract
The Sox family of transcription factors have been widely studied in the context of oligodendrocyte development. However, comparatively little is known about the role of Sox2, especially during CNS remyelination. Here we show that the expression of Sox2 occurs in oligodendrocyte progenitor cells (OPCs) in rodent models during myelination and in activated adult OPCs responding to demyelination, and is also detected in multiple sclerosis lesions. In normal adult white matter of both mice and rats, it is neither expressed by adult OPCs nor by oligodendrocytes (although it is expressed by a subpopulation of adult astrocytes). Overexpression of Sox2 in rat OPCs in vitro maintains the cells in a proliferative state and inhibits differentiation, while Sox2 knockout results in decreased OPC proliferation and survival, suggesting that Sox2 contributes to the expansion of OPCs during the recruitment phase of remyelination. Loss of function in cultured mouse OPCs also results in an impaired ability to undergo normal differentiation in response to differentiation signals, suggesting that Sox2 expression in activated OPCs also primes these cells to eventually undergo differentiation. In vivo studies on remyelination following experimental toxin-induced demyelination in mice with inducible loss of Sox2 revealed impaired remyelination, which was largely due to a profound attenuation of OPC recruitment and likely also due to impaired differentiation. Our results reveal a key role of Sox2 expression in OPCs responding to demyelination, enabling them to effectively contribute to remyelination. SIGNIFICANCE STATEMENT Understanding the mechanisms of CNS remyelination is central to developing effective means by which this process can be therapeutically enhanced in chronic demyelinating diseases such as multiple sclerosis. In this study, we describe the role of Sox2, a transcription factor widely implicated in stem cell biology, in CNS myelination and remyelination. We show how Sox2 is expressed in oligodendrocyte progenitor cells (OPCs) preparing to undergo differentiation, allowing them to undergo proliferation and priming them for subsequent differentiation. Although Sox2 is unlikely to be a direct therapeutic target, these data nevertheless provide more information on how OPC differentiation is controlled and therefore enriches our understanding of this important CNS regenerative process.
Collapse
Affiliation(s)
- Chao Zhao
- Wellcome Trust-Medical Research Council Stem Cell Institute and Department of Clinical Neurosciences, University of Cambridge, Cambridge Biomedical Campus, Cambridge CB2 0AH, United Kingdom, and
| | - Dan Ma
- Wellcome Trust-Medical Research Council Stem Cell Institute and Department of Clinical Neurosciences, University of Cambridge, Cambridge Biomedical Campus, Cambridge CB2 0AH, United Kingdom, and
| | - Malgorzata Zawadzka
- Wellcome Trust-Medical Research Council Stem Cell Institute and Department of Clinical Neurosciences, University of Cambridge, Cambridge Biomedical Campus, Cambridge CB2 0AH, United Kingdom, and
| | - Stephen P J Fancy
- Wellcome Trust-Medical Research Council Stem Cell Institute and Department of Clinical Neurosciences, University of Cambridge, Cambridge Biomedical Campus, Cambridge CB2 0AH, United Kingdom, and
| | - Lowri Elis-Williams
- Wellcome Trust-Medical Research Council Stem Cell Institute and Department of Clinical Neurosciences, University of Cambridge, Cambridge Biomedical Campus, Cambridge CB2 0AH, United Kingdom, and
| | - Guy Bouvier
- Wellcome Trust-Medical Research Council Stem Cell Institute and Department of Clinical Neurosciences, University of Cambridge, Cambridge Biomedical Campus, Cambridge CB2 0AH, United Kingdom, and
| | - John H Stockley
- Wellcome Trust-Medical Research Council Stem Cell Institute and Department of Clinical Neurosciences, University of Cambridge, Cambridge Biomedical Campus, Cambridge CB2 0AH, United Kingdom, and
| | - Glaucia Monteiro de Castro
- Wellcome Trust-Medical Research Council Stem Cell Institute and Department of Clinical Neurosciences, University of Cambridge, Cambridge Biomedical Campus, Cambridge CB2 0AH, United Kingdom, and
| | - Bowei Wang
- Wellcome Trust-Medical Research Council Stem Cell Institute and Department of Clinical Neurosciences, University of Cambridge, Cambridge Biomedical Campus, Cambridge CB2 0AH, United Kingdom, and
| | - Sabrina Jacobs
- Wellcome Trust-Medical Research Council Stem Cell Institute and Department of Clinical Neurosciences, University of Cambridge, Cambridge Biomedical Campus, Cambridge CB2 0AH, United Kingdom, and
| | - Patrizia Casaccia
- Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York 10029-6574
| | - Robin J M Franklin
- Wellcome Trust-Medical Research Council Stem Cell Institute and Department of Clinical Neurosciences, University of Cambridge, Cambridge Biomedical Campus, Cambridge CB2 0AH, United Kingdom, and
| |
Collapse
|