1
|
Kucenas S, Pulh P, Topilko P, Smith CJ. Glia at Transition Zones. Cold Spring Harb Perspect Biol 2025; 17:a041369. [PMID: 38858073 PMCID: PMC11864109 DOI: 10.1101/cshperspect.a041369] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/12/2024]
Abstract
Neural cells are segregated into their distinct central nervous system (CNS) and peripheral nervous system (PNS) domains. However, at specialized regions of the nervous system known as transition zones (TZs), glial cells from both the CNS and PNS are uniquely present with other specialized TZ cells. Herein we review the current understanding of vertebrate TZ cells. The article discusses the distinct cells at vertebrate TZs with a focus on cells that are located on the peripheral side of the spinal cord TZs. In addition to the developmental origin and differentiation of these TZ cells, the functional importance and the role of TZ cells in disease are highlighted. This article also reviews the common and unique features of vertebrate TZs from zebrafish to mice. We propose challenges and open questions in the field that could lead to exciting insights in the field of glial biology.
Collapse
Affiliation(s)
- Sarah Kucenas
- Department of Biology, University of Virginia, Charlottesville, Virginia 22904, USA
| | - Pernelle Pulh
- Institut Mondor de Recherche Biomédicale, Inserm U955-Team 9, 94010 Créteil, France
| | - Piotr Topilko
- Institut Mondor de Recherche Biomédicale, Inserm U955-Team 9, 94010 Créteil, France
| | - Cody J Smith
- Department of Biological Sciences, University of Notre Dame, Notre Dame, Indiana 46556, USA
| |
Collapse
|
2
|
Zeger L, Barasa P, Han Y, Hellgren J, Redwan IN, Reiche ME, Florin G, Christoffersson G, Kozlova EN. Microgravity Effect on Pancreatic Islets. Cells 2024; 13:1588. [PMID: 39329769 PMCID: PMC11430520 DOI: 10.3390/cells13181588] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Revised: 09/05/2024] [Accepted: 09/19/2024] [Indexed: 09/28/2024] Open
Abstract
We previously demonstrated that boundary cap neural crest stem cells (BCs) induce the proliferation of beta-cells in vitro, increase survival of pancreatic islets (PIs) in vivo after transplantation, and themselves strongly increase their proliferation capacity after exposure to space conditions. Therefore, we asked if space conditions can induce the proliferation of beta-cells when PIs are alone or together with BCs in free-floating or 3D-printed form. During the MASER 15 sounding rocket experiment, half of the cells were exposed to 6 min of microgravity (µg), whereas another group of cells were kept in 1 g conditions in a centrifuge onboard. The proliferation marker EdU was added to the cells just before the rocket reached µg conditions. The morphological assessment revealed that PIs successfully survived and strongly proliferated, particularly in the free-floating condition, though the fusion of PIs hampered statistical analysis. Proliferation of beta-cells was displayed in 3D-printed islets two weeks after µg exposure, suggesting that the effects of µg may be delayed. Thus, PIs in 3D-printed scaffolds did not fuse, and this preparation is more suitable than free-floating specimens for morphological analysis in µg studies. PIs maintained their increased proliferation capacity for weeks after µg exposure, an effect that may not appear directly, but can emerge after a delay.
Collapse
Affiliation(s)
- Lukas Zeger
- Regenerative Neurobiology, Department of Immunology, Genetics and Pathology, Uppsala University, 75108 Uppsala, Sweden; (L.Z.); (Y.H.)
| | - Povilas Barasa
- Department of Biological Models, Institute of Biochemistry, Vilnius University, LT-08662 Vilnius, Lithuania;
| | - Yilin Han
- Regenerative Neurobiology, Department of Immunology, Genetics and Pathology, Uppsala University, 75108 Uppsala, Sweden; (L.Z.); (Y.H.)
| | - Josefin Hellgren
- CELLINK Bioprinting AB, Langfilsgatan 7, 41277 Gothenburg, Sweden; (J.H.); (I.N.R.)
| | - Itedale Namro Redwan
- CELLINK Bioprinting AB, Langfilsgatan 7, 41277 Gothenburg, Sweden; (J.H.); (I.N.R.)
| | - Myrthe E. Reiche
- Department of Medical Cell Biology, Science for Life Laboratory, Uppsala University, 75310 Uppsala, Sweden; (M.E.R.); (G.C.)
| | | | - Gustaf Christoffersson
- Department of Medical Cell Biology, Science for Life Laboratory, Uppsala University, 75310 Uppsala, Sweden; (M.E.R.); (G.C.)
| | - Elena N. Kozlova
- Regenerative Neurobiology, Department of Immunology, Genetics and Pathology, Uppsala University, 75108 Uppsala, Sweden; (L.Z.); (Y.H.)
| |
Collapse
|
3
|
Han Y, Barasa P, Zeger L, Salomonsson SB, Zanotti F, Egli M, Zavan B, Trentini M, Florin G, Vaerneus A, Aldskogius H, Fredriksson R, Kozlova EN. Effects of microgravity on neural crest stem cells. Front Neurosci 2024; 18:1379076. [PMID: 38660221 PMCID: PMC11041629 DOI: 10.3389/fnins.2024.1379076] [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: 01/30/2024] [Accepted: 03/15/2024] [Indexed: 04/26/2024] Open
Abstract
Exposure to microgravity (μg) results in a range of systemic changes in the organism, but may also have beneficial cellular effects. In a previous study we detected increased proliferation capacity and upregulation of genes related to proliferation and survival in boundary cap neural crest stem cells (BC) after MASER14 sounding rocket flight compared to ground-based controls. However, whether these changes were due to μg or hypergravity was not clarified. In the current MASER15 experiment BCs were exposed simultaneously to μg and 1 g conditions provided by an onboard centrifuge. BCs exposed to μg displayed a markedly increased proliferation capacity compared to 1 g on board controls, and genetic analysis of BCs harvested 5 h after flight revealed an upregulation, specifically in μg-exposed BCs, of Zfp462 transcription factor, a key regulator of cell pluripotency and neuronal fate. This was associated with alterations in exosome microRNA content between μg and 1 g exposed MASER15 specimens. Since the specimens from MASER14 were obtained for analysis with 1 week's delay, we examined whether gene expression and exosome content were different compared to the current MASER15 experiments, in which specimens were harvested 5 h after flight. The overall pattern of gene expression was different and Zfp462 expression was down-regulated in MASER14 BC μg compared to directly harvested specimens (MASER15). MicroRNA exosome content was markedly altered in medium harvested with delay compared to directly collected samples. In conclusion, our analysis indicates that even short exposure to μg alters gene expression, leading to increased BC capacity for proliferation and survival, lasting for a long time after μg exposure. With delayed harvest of specimens, a situation which may occur due to special post-flight circumstances, the exosome microRNA content is modified compared to fast specimen harvest, and the direct effects from μg exposure may be partially attenuated, whereas other effects can last for a long time after return to ground conditions.
Collapse
Affiliation(s)
- Yilin Han
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Povilas Barasa
- Institute of Biochemistry, Vilnius University, Vilnius, Lithuania
| | - Lukas Zeger
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Sara B. Salomonsson
- Department of Pharmaceutical Bioscience, Uppsala University, Uppsala, Sweden
| | - Federica Zanotti
- Department of Translational Medicine, University of Ferrara, Ferrara, Italy
| | - Marcel Egli
- Space Biology Group, School of Engineering and Architecture, Institute of Medical Engineering, Lucerne University of Applied Sciences and Arts, Hergiswil, Switzerland
- National Center for Biomedical Research in Space, Innovation Cluster Space and Aviation, University of Zurich, Zurich, Switzerland
| | - Barbara Zavan
- Department of Translational Medicine, University of Ferrara, Ferrara, Italy
| | - Martina Trentini
- Department of Translational Medicine, University of Ferrara, Ferrara, Italy
| | | | | | - Håkan Aldskogius
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Robert Fredriksson
- Department of Pharmaceutical Bioscience, Uppsala University, Uppsala, Sweden
| | - Elena N. Kozlova
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| |
Collapse
|
4
|
Staedtke V, Anstett K, Bedwell D, Giovannini M, Keeling K, Kesterson R, Kim Y, Korf B, Leier A, McManus ML, Sarnoff H, Vitte J, Walker JA, Plotkin SR, Wallis D. Gene-targeted therapy for neurofibromatosis and schwannomatosis: The path to clinical trials. Clin Trials 2024; 21:51-66. [PMID: 37937606 DOI: 10.1177/17407745231207970] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2023]
Abstract
Numerous successful gene-targeted therapies are arising for the treatment of a variety of rare diseases. At the same time, current treatment options for neurofibromatosis 1 and schwannomatosis are limited and do not directly address loss of gene/protein function. In addition, treatments have mostly focused on symptomatic tumors, but have failed to address multisystem involvement in these conditions. Gene-targeted therapies hold promise to address these limitations. However, despite intense interest over decades, multiple preclinical and clinical issues need to be resolved before they become a reality. The optimal approaches to gene-, mRNA-, or protein restoration and to delivery to the appropriate cell types remain elusive. Preclinical models that recapitulate manifestations of neurofibromatosis 1 and schwannomatosis need to be refined. The development of validated assays for measuring neurofibromin and merlin activity in animal and human tissues will be critical for early-stage trials, as will the selection of appropriate patients, based on their individual genotypes and risk/benefit balance. Once the safety of gene-targeted therapy for symptomatic tumors has been established, the possibility of addressing a wide range of symptoms, including non-tumor manifestations, should be explored. As preclinical efforts are underway, it will be essential to educate both clinicians and those affected by neurofibromatosis 1/schwannomatosis about the risks and benefits of gene-targeted therapy for these conditions.
Collapse
Affiliation(s)
- Verena Staedtke
- Department of Neurology, Johns Hopkins University, Baltimore, MD, USA
| | - Kara Anstett
- Department of Neurology, NYU Grossman School of Medicine, New York, NY, USA
| | - David Bedwell
- Department of Biochemistry and Molecular Genetics, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Marco Giovannini
- Department of Head and Neck Surgery, David Geffen School of Medicine at UCLA and Jonsson Comprehensive Cancer Center (JCCC), University of California Los Angeles, Los Angeles, CA, USA
| | - Kim Keeling
- Department of Biochemistry and Molecular Genetics, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Robert Kesterson
- Department of Cancer Precision Medicine, Pennington Biomedical Research Center, Baton Rouge, LA, USA
| | - YooRi Kim
- Gilbert Family Foundation, Detroit, MI, USA
| | - Bruce Korf
- Department of Genetics, The University of Alabama at Birmingham, Birmingham, AL, USA
| | - André Leier
- Department of Genetics, The University of Alabama at Birmingham, Birmingham, AL, USA
| | | | | | - Jeremie Vitte
- Department of Head and Neck Surgery, David Geffen School of Medicine at UCLA and Jonsson Comprehensive Cancer Center (JCCC), University of California Los Angeles, Los Angeles, CA, USA
| | - James A Walker
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Scott R Plotkin
- Department of Neurology and Cancer Center, Massachusetts General Hospital, Boston, MA, USA
| | - Deeann Wallis
- Department of Genetics, The University of Alabama at Birmingham, Birmingham, AL, USA
| |
Collapse
|
5
|
Fontenas L. Glial plasticity at nervous system transition zones. Biol Open 2023; 12:bio060037. [PMID: 37787575 PMCID: PMC10562931 DOI: 10.1242/bio.060037] [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: 10/04/2023] Open
Abstract
The central and peripheral nervous systems (CNS and PNS, respectively) are two separate yet connected domains characterized by molecularly distinct cellular components that communicate via specialized structures called transition zones to allow information to travel from the CNS to the periphery, and vice versa. Until recently, nervous system transition zones were thought to be selectively permeable only to axons, and the establishment of the territories occupied by glial cells at these complex regions remained poorly described and not well understood. Recent work now demonstrates that transition zones are occupied by dynamic glial cells and are precisely regulated over the course of nervous system development. This review highlights recent work on glial cell migration in and out of the spinal cord, at motor exit point (MEP) and dorsal root entry zone (DREZ) transition zones, in the physiological and diseased nervous systems. These cells include myelinating glia (oligodendrocyte lineage cells, Schwann cells and motor exit point glia), exit glia, perineurial cells that form the perineurium along spinal nerves, as well as professional and non-professional phagocytes (microglia and neural crest cells).
Collapse
Affiliation(s)
- Laura Fontenas
- Department of Biological Sciences, Florida Atlantic University, Jupiter, FL 33458, USA
| |
Collapse
|
6
|
Staedtke V, Topilko P, Le LQ, Grimes K, Largaespada DA, Cagan RL, Steensma MR, Stemmer-Rachamimov A, Blakeley JO, Rhodes SD, Ly I, Romo CG, Lee SY, Serra E. Existing and Developing Preclinical Models for Neurofibromatosis Type 1-Related Cutaneous Neurofibromas. J Invest Dermatol 2023; 143:1378-1387. [PMID: 37330719 PMCID: PMC11246562 DOI: 10.1016/j.jid.2023.01.042] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Revised: 01/17/2023] [Accepted: 01/19/2023] [Indexed: 06/19/2023]
Abstract
Neurofibromatosis type 1 (NF1) is caused by a nonfunctional copy of the NF1 tumor suppressor gene that predisposes patients to the development of cutaneous neurofibromas (cNFs), the skin tumor that is the hallmark of this condition. Innumerable benign cNFs, each appearing by an independent somatic inactivation of the remaining functional NF1 allele, form in nearly all patients with NF1. One of the limitations in developing a treatment for cNFs is an incomplete understanding of the underlying pathophysiology and limitations in experimental modeling. Recent advances in preclinical in vitro and in vivo modeling have substantially enhanced our understanding of cNF biology and created unprecedented opportunities for therapeutic discovery. We discuss the current state of cNF preclinical in vitro and in vivo model systems, including two- and three-dimensional cell cultures, organoids, genetically engineered mice, patient-derived xenografts, and porcine models. We highlight the models' relationship to human cNFs and how they can be used to gain insight into cNF development and therapeutic discovery.
Collapse
Affiliation(s)
- Verena Staedtke
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.
| | - Piotr Topilko
- Institut Mondor de Recherche Biomédicale (IMRB), Créteil, France
| | - Lu Q Le
- Department of Dermatology, UT Southwestern Medical Center, Dallas, Texas, USA
| | - Kevin Grimes
- SPARK Program in Translational Research, Stanford University School of Medicine, Stanford, California, USA; Department of Chemical and Systems Biology, Stanford University School of Medicine, Stanford, California, USA
| | - David A Largaespada
- Department of Pediatrics, University of Minnesota, Minneapolis, Minnesota, USA
| | - Ross L Cagan
- School of Cancer Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Matthew R Steensma
- Center for Cancer and Cell Biology, Van Andel Research Institute, Grand Rapids, Michigan, USA; Helen DeVos Children's Hospital, Spectrum Health System, Grand Rapids, Michigan, USA; Michigan State University College of Human Medicine, Grand Rapids, Michigan, USA
| | - Anat Stemmer-Rachamimov
- Department of Pathology and Center for Cancer Research, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Jaishri O Blakeley
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Steven D Rhodes
- Division of Hematology-Oncology, Department of Pediatrics, Indiana University School of Medicine, Indianapolis, Indiana, USA; Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, Indiana, USA; Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, Indiana, USA; Indiana University Melvin and Bren Simon Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Ina Ly
- Stephen E. and Catherine Pappas Center for Neuro-Oncology, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Carlos G Romo
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Sang Y Lee
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Eduard Serra
- Hereditary Cancer Group, Germans Trias i Pujol Research Institute (IGTP), Can Ruti Campus, Barcelona, Spain; Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain
| |
Collapse
|
7
|
Rasia-Filho AA, Calcagnotto ME, von Bohlen Und Halbach O. Glial Cell Modulation of Dendritic Spine Structure and Synaptic Function. ADVANCES IN NEUROBIOLOGY 2023; 34:255-310. [PMID: 37962798 DOI: 10.1007/978-3-031-36159-3_6] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
Glia comprise a heterogeneous group of cells involved in the structure and function of the central and peripheral nervous system. Glial cells are found from invertebrates to humans with morphological specializations related to the neural circuits in which they are embedded. Glial cells modulate neuronal functions, brain wiring and myelination, and information processing. For example, astrocytes send processes to the synaptic cleft, actively participate in the metabolism of neurotransmitters, and release gliotransmitters, whose multiple effects depend on the targeting cells. Human astrocytes are larger and more complex than their mice and rats counterparts. Astrocytes and microglia participate in the development and plasticity of neural circuits by modulating dendritic spines. Spines enhance neuronal connectivity, integrate most postsynaptic excitatory potentials, and balance the strength of each input. Not all central synapses are engulfed by astrocytic processes. When that relationship occurs, a different pattern for thin and large spines reflects an activity-dependent remodeling of motile astrocytic processes around presynaptic and postsynaptic elements. Microglia are equally relevant for synaptic processing, and both glial cells modulate the switch of neuroendocrine secretion and behavioral display needed for reproduction. In this chapter, we provide an overview of the structure, function, and plasticity of glial cells and relate them to synaptic maturation and modulation, also involving neurotrophic factors. Together, neurons and glia coordinate synaptic transmission in both normal and abnormal conditions. Neglected over decades, this exciting research field can unravel the complexity of species-specific neural cytoarchitecture as well as the dynamic region-specific functional interactions between diverse neurons and glial subtypes.
Collapse
Affiliation(s)
- Alberto A Rasia-Filho
- Department of Basic Sciences/Physiology and Graduate Program in Biosciences, Universidade Federal de Ciências da Saúde de Porto Alegre, Porto Alegre, RS, Brazil
- Graduate Program in Neuroscience, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Maria Elisa Calcagnotto
- Graduate Program in Neuroscience, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
- Department of Biochemistry, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
- Graduate Program in Biochemistry, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
- Graduate Program in Psychiatry and Behavioral Science, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | | |
Collapse
|
8
|
Borda M, Aquino JB, Mazzone GL. Cell-based experimental strategies for myelin repair in multiple sclerosis. J Neurosci Res 2023; 101:86-111. [PMID: 36164729 DOI: 10.1002/jnr.25129] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2022] [Revised: 08/21/2022] [Accepted: 09/09/2022] [Indexed: 11/10/2022]
Abstract
Multiple sclerosis (MS) is an autoimmune demyelinating disorder of the central nervous system (CNS), diagnosed at a mean age of 32 years. CNS glia are crucial players in the onset of MS, primarily involving astrocytes and microglia that can cause/allow massive oligodendroglial cells death, without immune cell infiltration. Current therapeutic approaches are aimed at modulating inflammatory reactions during relapsing episodes, but lack the ability to induce very significant repair mechanisms. In this review article, different experimental approaches based mainly on the application of different cell types as therapeutic strategies applied for the induction of myelin repair and/or the amelioration of the disease are discussed. Regarding this issue, different cell sources were applied in various experimental models of MS, with different results, both in significant improvements in remyelination and the reduction of neuroinflammation and glial activation, or in neuroprotection. All cell types tested have advantages and disadvantages, which makes it difficult to choose a better option for therapeutic application in MS. New strategies combining cell-based treatment with other applications would result in further improvements and would be good candidates for MS cell therapy and myelin repair.
Collapse
Affiliation(s)
- Maximiliano Borda
- Instituto de Investigaciones en Medicina Traslacional (IIMT), CONICET-Universidad Austral, Derqui, Pilar, Buenos Aires, Argentina
| | - Jorge B Aquino
- Instituto de Investigaciones en Medicina Traslacional (IIMT), CONICET-Universidad Austral, Derqui, Pilar, Buenos Aires, Argentina.,CONICET, Comisión Nacional de Investigaciones Científicas y Técnicas
| | - Graciela L Mazzone
- Instituto de Investigaciones en Medicina Traslacional (IIMT), CONICET-Universidad Austral, Derqui, Pilar, Buenos Aires, Argentina.,CONICET, Comisión Nacional de Investigaciones Científicas y Técnicas
| |
Collapse
|
9
|
Schwann cell functions in peripheral nerve development and repair. Neurobiol Dis 2023; 176:105952. [PMID: 36493976 DOI: 10.1016/j.nbd.2022.105952] [Citation(s) in RCA: 83] [Impact Index Per Article: 41.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Revised: 11/23/2022] [Accepted: 12/05/2022] [Indexed: 12/12/2022] Open
Abstract
The glial cell of the peripheral nervous system (PNS), the Schwann cell (SC), counts among the most multifaceted cells of the body. During development, SCs secure neuronal survival and participate in axonal path finding. Simultaneously, they orchestrate the architectural set up of the developing nerves, including the blood vessels and the endo-, peri- and epineurial layers. Perinatally, in rodents, SCs radially sort and subsequently myelinate individual axons larger than 1 μm in diameter, while small calibre axons become organised in non-myelinating Remak bundles. SCs have a vital role in maintaining axonal health throughout life and several specialized SC types perform essential functions at specific locations, such as terminal SC at the neuromuscular junction (NMJ) or SC within cutaneous sensory end organs. In addition, neural crest derived satellite glia maintain a tight communication with the soma of sensory, sympathetic, and parasympathetic neurons and neural crest derivatives are furthermore an indispensable part of the enteric nervous system. The remarkable plasticity of SCs becomes evident in the context of a nerve injury, where SC transdifferentiate into intriguing repair cells, which orchestrate a regenerative response that promotes nerve repair. Indeed, the multiple adaptations of SCs are captivating, but remain often ill-resolved on the molecular level. Here, we summarize and discuss the knowns and unknowns of the vast array of functions that this single cell type can cover in peripheral nervous system development, maintenance, and repair.
Collapse
|
10
|
Reed CB, Feltri ML, Wilson ER. Peripheral glia diversity. J Anat 2022; 241:1219-1234. [PMID: 34131911 PMCID: PMC8671569 DOI: 10.1111/joa.13484] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Revised: 05/20/2021] [Accepted: 05/26/2021] [Indexed: 12/13/2022] Open
Abstract
Recent years have seen an evolving appreciation for the role of glial cells in the nervous system. As we move away from the typical neurocentric view of neuroscience, the complexity and variability of central nervous system glia is emerging, far beyond the three main subtypes: astrocytes, oligodendrocytes, and microglia. Yet the diversity of the glia found in the peripheral nervous system remains rarely discussed. In this review, we discuss the developmental origin, morphology, and function of the different populations of glia found in the peripheral nervous system, including: myelinating Schwann cells, Remak Schwann cells, repair Schwann cells, satellite glia, boundary cap-derived glia, perineurial glia, terminal Schwann cells, glia found in the skin, olfactory ensheathing cells, and enteric glia. The morphological and functional heterogeneity of glia found in the periphery reflects the diverse roles the nervous system performs throughout the body. Further, it highlights a complexity that should be appreciated and considered when it comes to a complete understanding of the peripheral nervous system in health and disease.
Collapse
Affiliation(s)
- Chelsey B. Reed
- Hunter James Kelly Research InstituteJacobs School of Medicine and Biomedical Sciences StateUniversity of New York at BuffaloBuffaloNew YorkUSA
- Department of NeurologyJacobs School of Medicine and Biomedical SciencesState University of New York at BuffaloBuffaloNew YorkUSA
| | - M. Laura Feltri
- Hunter James Kelly Research InstituteJacobs School of Medicine and Biomedical Sciences StateUniversity of New York at BuffaloBuffaloNew YorkUSA
- Department of NeurologyJacobs School of Medicine and Biomedical SciencesState University of New York at BuffaloBuffaloNew YorkUSA
- Department of BiochemistryJacobs School of Medicine and Biomedical SciencesState University of New York at BuffaloBuffaloNew YorkUSA
| | - Emma R. Wilson
- Hunter James Kelly Research InstituteJacobs School of Medicine and Biomedical Sciences StateUniversity of New York at BuffaloBuffaloNew YorkUSA
- Department of BiochemistryJacobs School of Medicine and Biomedical SciencesState University of New York at BuffaloBuffaloNew YorkUSA
| |
Collapse
|
11
|
Ge LL, Xing MY, Zhang HB, Wang ZC. Neurofibroma Development in Neurofibromatosis Type 1: Insights from Cellular Origin and Schwann Cell Lineage Development. Cancers (Basel) 2022; 14:cancers14184513. [PMID: 36139671 PMCID: PMC9497298 DOI: 10.3390/cancers14184513] [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] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Revised: 09/11/2022] [Accepted: 09/14/2022] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND Neurofibromatosis type 1 (NF1), a genetic tumor predisposition syndrome that affects about 1 in 3000 newborns, is caused by mutations in the NF1 gene and subsequent inactivation of its encoded neurofibromin. Neurofibromin is a tumor suppressor protein involved in the downregulation of Ras signaling. Despite a diverse clinical spectrum, one of several hallmarks of NF1 is a peripheral nerve sheath tumor (PNST), which comprises mixed nervous and fibrous components. The distinct spatiotemporal characteristics of plexiform and cutaneous neurofibromas have prompted hypotheses about the origin and developmental features of these tumors, involving various cellular transition processes. METHODS We retrieved published literature from PubMed, EMBASE, and Web of Science up to 21 June 2022 and searched references cited in the selected studies to identify other relevant papers. Original articles reporting the pathogenesis of PNSTs during development were included in this review. We highlighted the Schwann cell (SC) lineage shift to better present the evolution of its corresponding cellular origin hypothesis and its important effects on the progression and malignant transformation of neurofibromas. CONCLUSIONS In this review, we summarized the vast array of evidence obtained on the full range of neurofibroma development based on cellular and molecular pathogenesis. By integrating findings relating to tumor formation, growth, and malignancy, we hope to reveal the role of SC lineage shift as well as the combined impact of additional determinants in the natural history of PNSTs.
Collapse
Affiliation(s)
- Ling-Ling Ge
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People′s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - Ming-Yan Xing
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200011, China
| | - Hai-Bing Zhang
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200011, China
- Correspondence: (H.-B.Z.); or (Z.-C.W.); Tel.: +86-021-54920988 (H.-B.Z.); +86-021-53315120 (Z.-C.W.)
| | - Zhi-Chao Wang
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People′s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
- Correspondence: (H.-B.Z.); or (Z.-C.W.); Tel.: +86-021-54920988 (H.-B.Z.); +86-021-53315120 (Z.-C.W.)
| |
Collapse
|
12
|
Abstract
Schwann cells in the peripheral nervous system (PNS) are essential for the support and myelination of axons, ensuring fast and accurate communication between the central nervous system and the periphery. Schwann cells and related glia accompany innervating axons in virtually all tissues in the body, where they exhibit remarkable plasticity and the ability to modulate pathology in extraordinary, and sometimes surprising, ways. Here, we provide a brief overview of the various glial cell types in the PNS and describe the cornerstone cellular and molecular processes that enable Schwann cells to perform their canonical functions. We then dive into discussing exciting noncanonical functions of Schwann cells and related PNS glia, which include their role in organizing the PNS, in regulating synaptic activity and pain, in modulating immunity, in providing a pool of stem cells for different organs, and, finally, in influencing cancer.
Collapse
Affiliation(s)
- Carla Taveggia
- Axo-Glial Interaction Unit, Division of Neuroscience, IRCCS Ospedale San Raffaele, Milan, Italy;
| | - M. Laura Feltri
- Institute for Myelin and Glia Exploration, Departments of Biochemistry and Neurology, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, New York, USA
| |
Collapse
|
13
|
Towards 3D Bioprinted Spinal Cord Organoids. Int J Mol Sci 2022; 23:ijms23105788. [PMID: 35628601 PMCID: PMC9144715 DOI: 10.3390/ijms23105788] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Revised: 05/16/2022] [Accepted: 05/18/2022] [Indexed: 12/14/2022] Open
Abstract
Three-dimensional (3D) cultures, so-called organoids, have emerged as an attractive tool for disease modeling and therapeutic innovations. Here, we aim to determine if boundary cap neural crest stem cells (BC) can survive and differentiate in gelatin-based 3D bioprinted bioink scaffolds in order to establish an enabling technology for the fabrication of spinal cord organoids on a chip. BC previously demonstrated the ability to support survival and differentiation of co-implanted or co-cultured cells and supported motor neuron survival in excitotoxically challenged spinal cord slice cultures. We tested different combinations of bioink and cross-linked material, analyzed the survival of BC on the surface and inside the scaffolds, and then tested if human iPSC-derived neural cells (motor neuron precursors and astrocytes) can be printed with the same protocol, which was developed for BC. We showed that this protocol is applicable for human cells. Neural differentiation was more prominent in the peripheral compared to central parts of the printed construct, presumably because of easier access to differentiation-promoting factors in the medium. These findings show that the gelatin-based and enzymatically cross-linked hydrogel is a suitable bioink for building a multicellular, bioprinted spinal cord organoid, but that further measures are still required to achieve uniform neural differentiation.
Collapse
|
14
|
Liu Y, Zhou S, Zhao L, Gu X. Identification of Neuronal Cells in Sciatic Nerves of Adult Rats. Front Cell Neurosci 2022; 16:816814. [PMID: 35401123 PMCID: PMC8991689 DOI: 10.3389/fncel.2022.816814] [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: 11/25/2021] [Accepted: 02/14/2022] [Indexed: 11/13/2022] Open
Abstract
Prior research generally confirms that there are no neuronal cell bodies in the adult sciatic nerve. However, we occasionally find some neuronal cells in adult rat sciatic nerves, either intact or crush-injured. By whole-mount staining and optical imaging of the hyalinized sciatic nerves for Stmn2 (a specific marker for neuronal cells), we found those neuronal cells with irregular distribution in the sciatic nerves in both crushed model and normal rats. We investigated the identity of those cells and established a cultured sciatic nerve model. Immunohistochemistry evidence both in vivo and in vitro illustrated that some of those cells are mature neurons in sciatic nerves. With single-cell sequencing of neuronal cells in adeno-associated virus (AAV)-infected sciatic nerves, we identified that some of those cells are a kind of neuronal stem-like cells. Then we constructed a Nestin-CreERT 2 rat line and traced those cells with fluorescence labeling which was induced by tamoxifen. Interesting, we proved that neuronal stem-like cells could proliferate by combination of EdU incorporation with staining in the sciatic nerves of transgenic rats. Together, the discovery of neuronal cells in adult sciatic nerves will make us aware of the distribution of neurons in the peripheral nervous system. Especially our data suggest that neuronal stem-like cells could proliferate in the sciatic nerves of adult rats.
Collapse
Affiliation(s)
- Yisheng Liu
- Model Animal Research Center, Nanjing University, Nanjing, China
| | - Songlin Zhou
- Key Laboratory of Neuroregeneration, Ministry of Education and Jiangsu Province, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China
| | - Lili Zhao
- Model Animal Research Center, Nanjing University, Nanjing, China
| | - Xiaosong Gu
- Model Animal Research Center, Nanjing University, Nanjing, China
- Key Laboratory of Neuroregeneration, Ministry of Education and Jiangsu Province, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China
| |
Collapse
|
15
|
Sinegubov A, Andreeva D, Burzak N, Vasyutina M, Murashova L, Dyachuk V. Heterogeneity and Potency of Peripheral Glial Cells in Embryonic Development and Adults. Front Mol Neurosci 2022; 15:737949. [PMID: 35401107 PMCID: PMC8990813 DOI: 10.3389/fnmol.2022.737949] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Accepted: 02/08/2022] [Indexed: 11/13/2022] Open
Abstract
This review describes the heterogeneity of peripheral glial cell populations, from the emergence of Schwann cells (SCs) in early development, to their involvement, and that of their derivatives in adult glial populations. We focus on the origin of the first glial precursors from neural crest cells (NCCs), and their ability to differentiate into several cell types during development. We also discuss the heterogeneity of embryonic glia in light of the latest data from genetic tracing and transcriptome analysis. Special attention has been paid to the biology of glial populations in adult animals, by highlighting common features of different glial cell types and molecular differences that modulate their functions. Finally, we consider the communication of glial cells with axons of neurons in normal and pathological conditions. In conclusion, the present review details how information available on glial cell types and their functions in normal and pathological conditions may be utilized in the development of novel therapeutic strategies for the treatment of patients with neurodiseases.
Collapse
|
16
|
Modeling iPSC-derived human neurofibroma-like tumors in mice uncovers the heterogeneity of Schwann cells within plexiform neurofibromas. Cell Rep 2022; 38:110385. [PMID: 35172160 DOI: 10.1016/j.celrep.2022.110385] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Revised: 11/04/2021] [Accepted: 01/24/2022] [Indexed: 12/20/2022] Open
Abstract
Plexiform neurofibromas (pNFs) are developmental tumors that appear in neurofibromatosis type 1 individuals, constituting a major source of morbidity and potentially transforming into a highly metastatic sarcoma (MPNST). pNFs arise after NF1 inactivation in a cell of the neural crest (NC)-Schwann cell (SC) lineage. Here, we develop an iPSC-based NC-SC in vitro differentiation system and construct a lineage expression roadmap for the analysis of different 2D and 3D NF models. The best model consists of generating heterotypic spheroids (neurofibromaspheres) composed of iPSC-derived differentiating NF1(-/-) SCs and NF1(+/-) pNF-derived fibroblasts (Fbs). Neurofibromaspheres form by maintaining highly proliferative NF1(-/-) cells committed to the NC-SC axis due to SC-SC and SC-Fb interactions, resulting in SC linage cells at different maturation points. Upon engraftment on the mouse sciatic nerve, neurofibromaspheres consistently generate human NF-like tumors. Analysis of expression roadmap genes in human pNF single-cell RNA-seq data uncovers the presence of SC subpopulations at distinct differentiation states.
Collapse
|
17
|
Kolos EA, Korzhevskii DE. Glutamine Synthetase in the Cells of the Developing Rat Spinal Cord. Russ J Dev Biol 2021. [DOI: 10.1134/s1062360421050040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
18
|
Solovieva T, Bronner M. Reprint of: Schwann cell precursors: Where they come from and where they go. Cells Dev 2021; 168:203729. [PMID: 34456178 DOI: 10.1016/j.cdev.2021.203729] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Revised: 04/29/2021] [Accepted: 04/30/2021] [Indexed: 10/20/2022]
Abstract
Schwann cell precursors (SCPs) are a transient population in the embryo, closely associated with nerves along which they migrate into the periphery of the body. Long considered to be progenitors that only form Schwann cells-the myelinating cells of nerves, current evidence suggests that SCPs have much broader developmental potential. Indeed, different cell marking techniques employed over the past 20 years have identified multiple novel SCP derivatives throughout the body. It is now clear that SCPs represent a multipotent progenitor population, which also display a level of plasticity in response to injury. Moreover, they originate from multiple origins in the embryo and may reflect several distinct subpopulations in terms of molecular identity and fate. Here we review SCP origins, derivatives and plasticity in development, growth and repair.
Collapse
Affiliation(s)
- Tatiana Solovieva
- Division of Biology and Biological Engineering, California Institute of Technology, United States of America.
| | - Marianne Bronner
- Division of Biology and Biological Engineering, California Institute of Technology, United States of America
| |
Collapse
|
19
|
Han Y, Zeger L, Tripathi R, Egli M, Ille F, Lockowandt C, Florin G, Atic E, Redwan IN, Fredriksson R, Kozlova EN. Molecular genetic analysis of neural stem cells after space flight and simulated microgravity on earth. Biotechnol Bioeng 2021; 118:3832-3846. [PMID: 34125436 DOI: 10.1002/bit.27858] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Revised: 06/09/2021] [Accepted: 06/09/2021] [Indexed: 02/06/2023]
Abstract
Understanding how stem cells adapt to space flight conditions is fundamental for human space missions and extraterrestrial settlement. We analyzed gene expression in boundary cap neural crest stem cells (BCs), which are attractive for regenerative medicine by their ability to promote proliferation and survival of cocultured and co-implanted cells. BCs were launched to space (space exposed cells) (SEC), onboard sounding rocket MASER 14 as free-floating neurospheres or in a bioprinted scaffold. For comparison, BCs were placed in a random positioning machine (RPM) to simulate microgravity on earth (RPM cells) or were cultured under control conditions in the laboratory. Using next-generation RNA sequencing and data post-processing, we discovered that SEC upregulated genes related to proliferation and survival, whereas RPM cells upregulated genes associated with differentiation and inflammation. Thus, (i) space flight provides unique conditions with distinctly different effects on the properties of BC compared to earth controls, and (ii) the space flight exposure induces postflight properties that reinforce the utility of BC for regenerative medicine and tissue engineering.
Collapse
Affiliation(s)
- Yilin Han
- Department of Neuroscience, Regenerative Neurobiology, Uppsala University, Uppsala, Sweden
| | - Lukas Zeger
- Department of Neuroscience, Regenerative Neurobiology, Uppsala University, Uppsala, Sweden
| | - Rekha Tripathi
- Department of Pharmaceutical Bioscience, Molecular Pharmacology, Uppsala University, Uppsala, Sweden
| | - Marcel Egli
- Luzerne School of Engineering and Architecture, Institute of Medical Engineering (IMT), Luzerne, Switzerland
| | - Fabian Ille
- Luzerne School of Engineering and Architecture, Institute of Medical Engineering (IMT), Luzerne, Switzerland
| | | | - Gunnar Florin
- Swedish Space Corporation, Science Service Division, Solna, Sweden
| | | | | | - Robert Fredriksson
- Department of Pharmaceutical Bioscience, Molecular Pharmacology, Uppsala University, Uppsala, Sweden
| | - Elena N Kozlova
- Department of Neuroscience, Regenerative Neurobiology, Uppsala University, Uppsala, Sweden
| |
Collapse
|
20
|
Solovieva T, Bronner M. Schwann cell precursors: Where they come from and where they go. Cells Dev 2021; 166:203686. [PMID: 33994354 DOI: 10.1016/j.cdev.2021.203686] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Revised: 04/29/2021] [Accepted: 04/30/2021] [Indexed: 11/30/2022]
Abstract
Schwann cell precursors (SCPs) are a transient population in the embryo, closely associated with nerves along which they migrate into the periphery of the body. Long considered to be progenitors that only form Schwann cells-the myelinating cells of nerves, current evidence suggests that SCPs have much broader developmental potential. Indeed, different cell marking techniques employed over the past 20 years have identified multiple novel SCP derivatives throughout the body. It is now clear that SCPs represent a multipotent progenitor population, which also display a level of plasticity in response to injury. Moreover, they originate from multiple origins in the embryo and may reflect several distinct subpopulations in terms of molecular identity and fate. Here we review SCP origins, derivatives and plasticity in development, growth and repair.
Collapse
Affiliation(s)
- Tatiana Solovieva
- Division of Biology and Biological Engineering, California Institute of Technology, United States of America.
| | - Marianne Bronner
- Division of Biology and Biological Engineering, California Institute of Technology, United States of America
| |
Collapse
|
21
|
Yusifov E, Dumoulin A, Stoeckli ET. Investigating Primary Cilia during Peripheral Nervous System Formation. Int J Mol Sci 2021; 22:3176. [PMID: 33804711 PMCID: PMC8003989 DOI: 10.3390/ijms22063176] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Revised: 03/16/2021] [Accepted: 03/16/2021] [Indexed: 12/22/2022] Open
Abstract
The primary cilium plays a pivotal role during the embryonic development of vertebrates. It acts as a somatic signaling hub for specific pathways, such as Sonic Hedgehog signaling. In humans, mutations in genes that cause dysregulation of ciliogenesis or ciliary function lead to severe developmental disorders called ciliopathies. Beyond its role in early morphogenesis, growing evidence points towards an essential function of the primary cilium in neural circuit formation in the central nervous system. However, very little is known about a potential role in the formation of the peripheral nervous system. Here, we investigate the presence of the primary cilium in neural crest cells and their derivatives in the trunk of developing chicken embryos in vivo. We found that neural crest cells, sensory neurons, and boundary cap cells all bear a primary cilium during key stages of early peripheral nervous system formation. Moreover, we describe differences in the ciliation of neuronal cultures of different populations from the peripheral and central nervous systems. Our results offer a framework for further in vivo and in vitro investigations on specific roles that the primary cilium might play during peripheral nervous system formation.
Collapse
Affiliation(s)
| | | | - Esther T. Stoeckli
- Department of Molecular Life Sciences and Neuroscience Center Zurich, University of Zurich, Winterthurerstrasse 190, 8057 Zurich, Switzerland; (E.Y.); (A.D.)
| |
Collapse
|
22
|
Feltri ML, Weaver MR, Belin S, Poitelon Y. The Hippo pathway: Horizons for innovative treatments of peripheral nerve diseases. J Peripher Nerv Syst 2021; 26:4-16. [PMID: 33449435 DOI: 10.1111/jns.12431] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Revised: 12/16/2020] [Accepted: 12/20/2020] [Indexed: 12/19/2022]
Abstract
Initially identified in Drosophila, the Hippo signaling pathway regulates how cells respond to their environment by controlling proliferation, migration and differentiation. Many recent studies have focused on characterizing Hippo pathway function and regulation in mammalian cells. Here, we present a brief overview of the major components of the Hippo pathway, as well as their regulation and function. We comprehensively review the studies that have contributed to our understanding of the Hippo pathway in the function of the peripheral nervous system and in peripheral nerve diseases. Finally, we discuss innovative approaches that aim to modulate Hippo pathway components in diseases of the peripheral nervous system.
Collapse
Affiliation(s)
- M Laura Feltri
- Hunter James Kelly Research Institute, Department of Biochemistry, Jacobs School of Medicine and Biomedical Sciences, Buffalo, New York, USA.,Department of Neurology, Jacobs School of Medicine and Biomedical Sciences, Buffalo, New York, USA
| | - Michael R Weaver
- Hunter James Kelly Research Institute, Department of Biochemistry, Jacobs School of Medicine and Biomedical Sciences, Buffalo, New York, USA
| | - Sophie Belin
- Department of Neuroscience and Experimental Therapeutics, Albany Medical College, Albany, New York, USA
| | - Yannick Poitelon
- Department of Neuroscience and Experimental Therapeutics, Albany Medical College, Albany, New York, USA
| |
Collapse
|
23
|
Perera SN, Williams RM, Lyne R, Stubbs O, Buehler DP, Sauka-Spengler T, Noda M, Micklem G, Southard-Smith EM, Baker CVH. Insights into olfactory ensheathing cell development from a laser-microdissection and transcriptome-profiling approach. Glia 2020; 68:2550-2584. [PMID: 32857879 PMCID: PMC7116175 DOI: 10.1002/glia.23870] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Revised: 05/23/2020] [Accepted: 05/27/2020] [Indexed: 12/14/2022]
Abstract
Olfactory ensheathing cells (OECs) are neural crest-derived glia that ensheath bundles of olfactory axons from their peripheral origins in the olfactory epithelium to their central targets in the olfactory bulb. We took an unbiased laser microdissection and differential RNA-seq approach, validated by in situ hybridization, to identify candidate molecular mechanisms underlying mouse OEC development and differences with the neural crest-derived Schwann cells developing on other peripheral nerves. We identified 25 novel markers for developing OECs in the olfactory mucosa and/or the olfactory nerve layer surrounding the olfactory bulb, of which 15 were OEC-specific (that is, not expressed by Schwann cells). One pan-OEC-specific gene, Ptprz1, encodes a receptor-like tyrosine phosphatase that blocks oligodendrocyte differentiation. Mutant analysis suggests Ptprz1 may also act as a brake on OEC differentiation, and that its loss disrupts olfactory axon targeting. Overall, our results provide new insights into OEC development and the diversification of neural crest-derived glia.
Collapse
Affiliation(s)
- Surangi N. Perera
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
| | - Ruth M. Williams
- MRC Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Rachel Lyne
- Department of Genetics, University of Cambridge, Cambridge, UK
| | - Oliver Stubbs
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
| | - Dennis P. Buehler
- Division of Genetic Medicine, Department of Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Tatjana Sauka-Spengler
- MRC Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Masaharu Noda
- Division of Molecular Neurobiology, National Institute for Basic Biology, Okazaki, Japan
| | - Gos Micklem
- Department of Genetics, University of Cambridge, Cambridge, UK
| | - E. Michelle Southard-Smith
- Division of Genetic Medicine, Department of Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Clare V. H. Baker
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
| |
Collapse
|
24
|
Modeling tumors of the peripheral nervous system associated with Neurofibromatosis type 1: Reprogramming plexiform neurofibroma cells. Stem Cell Res 2020; 49:102068. [PMID: 33160273 DOI: 10.1016/j.scr.2020.102068] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Revised: 10/16/2020] [Accepted: 10/20/2020] [Indexed: 11/24/2022] Open
Abstract
Plexiform neurofibromas (pNFs) are benign tumors of the peripheral nervous system (PNS) that can progress towards a deadly soft tissue sarcoma termed malignant peripheral nerve sheath tumor (MPNST). pNFs appear during development in the context of the genetic disease Neurofibromatosis type 1 (NF1) due to the complete loss of the NF1 tumor suppressor gene in a cell of the neural crest (NC) - Schwann cell (SC) axis of differentiation. NF1(-/-) cells from pNFs can be reprogrammed into induced pluripotent stem cells (iPSCs) that exhibit an increased proliferation rate and maintain full iPSC properties. Efficient protocols for iPSC differentiation towards NC and SC exist and thus NC cells can be efficiently obtained from NF1(-/-) iPSCs and further differentiated towards SCs. In this review, we will focus on the iPSC modeling of pNFs, including the reprogramming of primary pNF-derived cells, the properties of pNF-derived iPSCs, the capacity to differentiate towards the NC-SC lineage, and how well iPSC-derived NF1(-/-) SC spheroids recapitulate pNF-derived primary SCs. The potential uses of NF1(-/-) iPSCs in pNF modeling and a future outlook are discussed.
Collapse
|
25
|
Perera SN, Kerosuo L. On the road again: Establishment and maintenance of stemness in the neural crest from embryo to adulthood. STEM CELLS (DAYTON, OHIO) 2020; 39:7-25. [PMID: 33017496 PMCID: PMC7821161 DOI: 10.1002/stem.3283] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Revised: 09/08/2020] [Accepted: 09/11/2020] [Indexed: 12/22/2022]
Abstract
Unique to vertebrates, the neural crest (NC) is an embryonic stem cell population that contributes to a greatly expanding list of derivatives ranging from neurons and glia of the peripheral nervous system, facial cartilage and bone, pigment cells of the skin to secretory cells of the endocrine system. Here, we focus on what is specifically known about establishment and maintenance of NC stemness and ultimate fate commitment mechanisms, which could help explain its exceptionally high stem cell potential that exceeds the "rules set during gastrulation." In fact, recent discoveries have shed light on the existence of NC cells that coexpress commonly accepted pluripotency factors like Nanog, Oct4/PouV, and Klf4. The coexpression of pluripotency factors together with the exceptional array of diverse NC derivatives encouraged us to propose a new term "pleistopotent" (Greek for abundant, a substantial amount) to be used to reflect the uniqueness of the NC as compared to other post-gastrulation stem cell populations in the vertebrate body, and to differentiate them from multipotent lineage restricted stem cells. We also discuss studies related to the maintenance of NC stemness within the challenging context of being a transient and thus a constantly changing population of stem cells without a permanent niche. The discovery of the stem cell potential of Schwann cell precursors as well as multiple adult NC-derived stem cell reservoirs during the past decade has greatly increased our understanding of how NC cells contribute to tissues formed after its initial migration stage in young embryos.
Collapse
Affiliation(s)
- Surangi N Perera
- Neural Crest Development and Disease Unit, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, Maryland, USA
| | - Laura Kerosuo
- Neural Crest Development and Disease Unit, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, Maryland, USA
| |
Collapse
|
26
|
Abstract
Neurofibromatosis type I (NF1) is a debilitating inherited tumor syndrome affecting around 1 in 3000 people. Patients present with a variety of tumors caused by biallelic loss of the tumor suppressor neurofibromin (NF1), a negative regulator of Ras signaling. While the mechanism of tumor formation is similar in the majority of NF1 cases, the clinical spectrum of tumors can vary depending on spatiotemporal loss of heterozygosity of NF1 in cells derived from the neural crest during development. The hallmark lesions that give NF1 its namesake are neurofibromas, which are benign Schwann cell tumors composed of nervous and fibrous tissue. Neurofibromas can be found in the skin (cutaneous neurofibroma) or deeper in body near nerve plexuses (plexiform neurofibroma). While neurofibromas have been known to be Schwann cell tumors for many years, the exact timing and initiating cell has remained elusive. This has led to difficulties in developing animal models and successful therapies for NF1. A culmination of recent genetic studies has finally begun to shed light on the detailed cellular origins of neurofibromatosis. In this review, we will examine the hunt for neurofibroma tumor cells of origin through a historical lens, detailing the genetic systems used to delineate the source of plexiform and cutaneous neurofibromas. Through these novel findings, we can better understand the cellular, temporal, and developmental context during tumor initiation. By leveraging this data, we hope to uncover new therapeutic targets and mechanisms to treat NF1 patients.
Collapse
Affiliation(s)
- Stephen Li
- Department of Dermatology, University of Texas Southwestern Medical Center, Dallas.,Medical Scientist Training Program, University of Texas Southwestern Medical Center, Dallas.,Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas
| | - Zhiguo Chen
- Department of Dermatology, University of Texas Southwestern Medical Center, Dallas
| | - Lu Q Le
- Department of Dermatology, University of Texas Southwestern Medical Center, Dallas.,Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas.,Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas.,Neurofibromatosis Clinic, University of Texas Southwestern Medical Center, Dallas
| |
Collapse
|
27
|
Shea GK, Tai EW, Leung KH, Mung AK, Li MT, Tsui AY, Tam AK, Shum DK, Chan Y. Juxtacrine signalling via Notch and ErbB receptors in the switch to fate commitment of bone marrow‐derived Schwann cells. Eur J Neurosci 2020; 52:3306-3321. [DOI: 10.1111/ejn.14837] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2019] [Revised: 05/03/2020] [Accepted: 05/18/2020] [Indexed: 01/09/2023]
Affiliation(s)
- Graham Ka‐Hon Shea
- Department of Orthopaedics and Traumatology Li Ka Shing Faculty of Medicine The University of Hong Kong Hong Kong Hong Kong
| | - Evelyn Wing‐Yin Tai
- Department of Orthopaedics and Traumatology Li Ka Shing Faculty of Medicine The University of Hong Kong Hong Kong Hong Kong
- Li Ka Shing Faculty of Medicine School of Biomedical Sciences The University of Hong Kong Hong Kong Hong Kong
| | - Katherine Ho‐Yan Leung
- Li Ka Shing Faculty of Medicine School of Biomedical Sciences The University of Hong Kong Hong Kong Hong Kong
| | - Alan Kwan‐Long Mung
- Department of Orthopaedics and Traumatology Li Ka Shing Faculty of Medicine The University of Hong Kong Hong Kong Hong Kong
- Li Ka Shing Faculty of Medicine School of Biomedical Sciences The University of Hong Kong Hong Kong Hong Kong
| | - Maximilian Tak‐Sui Li
- Li Ka Shing Faculty of Medicine School of Biomedical Sciences The University of Hong Kong Hong Kong Hong Kong
- Research Centre of Heart, Brain, Hormone & Healthy Aging The University of Hong Kong Hong Kong Hong Kong
| | - Alex Yat‐Ping Tsui
- Li Ka Shing Faculty of Medicine School of Biomedical Sciences The University of Hong Kong Hong Kong Hong Kong
- Research Centre of Heart, Brain, Hormone & Healthy Aging The University of Hong Kong Hong Kong Hong Kong
| | - Anthony Kin‐Wai Tam
- Li Ka Shing Faculty of Medicine School of Biomedical Sciences The University of Hong Kong Hong Kong Hong Kong
- Research Centre of Heart, Brain, Hormone & Healthy Aging The University of Hong Kong Hong Kong Hong Kong
| | - Daisy Kwok‐Yan Shum
- Li Ka Shing Faculty of Medicine School of Biomedical Sciences The University of Hong Kong Hong Kong Hong Kong
- Research Centre of Heart, Brain, Hormone & Healthy Aging The University of Hong Kong Hong Kong Hong Kong
- State Key Laboratory of Brain and Cognitive Science The University of Hong Kong Hong Kong Hong Kong
| | - Ying‐Shing Chan
- Li Ka Shing Faculty of Medicine School of Biomedical Sciences The University of Hong Kong Hong Kong Hong Kong
- Research Centre of Heart, Brain, Hormone & Healthy Aging The University of Hong Kong Hong Kong Hong Kong
- State Key Laboratory of Brain and Cognitive Science The University of Hong Kong Hong Kong Hong Kong
| |
Collapse
|
28
|
Suter TACS, Jaworski A. Cell migration and axon guidance at the border between central and peripheral nervous system. Science 2020; 365:365/6456/eaaw8231. [PMID: 31467195 DOI: 10.1126/science.aaw8231] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Accepted: 07/22/2019] [Indexed: 12/13/2022]
Abstract
The central and peripheral nervous system (CNS and PNS, respectively) are composed of distinct neuronal and glial cell types with specialized functional properties. However, a small number of select cells traverse the CNS-PNS boundary and connect these two major subdivisions of the nervous system. This pattern of segregation and selective connectivity is established during embryonic development, when neurons and glia migrate to their destinations and axons project to their targets. Here, we provide an overview of the cellular and molecular mechanisms that control cell migration and axon guidance at the vertebrate CNS-PNS border. We highlight recent advances on how cell bodies and axons are instructed to either cross or respect this boundary, and present open questions concerning the development and plasticity of the CNS-PNS interface.
Collapse
Affiliation(s)
- Tracey A C S Suter
- Department of Neuroscience, Division of Biology and Medicine, Brown University, Providence, RI 02912, USA.,Robert J. and Nancy D. Carney Institute for Brain Science, Providence, RI 02912, USA
| | - Alexander Jaworski
- Department of Neuroscience, Division of Biology and Medicine, Brown University, Providence, RI 02912, USA. .,Robert J. and Nancy D. Carney Institute for Brain Science, Providence, RI 02912, USA
| |
Collapse
|
29
|
Han Y, Baltriukienė D, Kozlova EN. Effect of scaffold properties on adhesion and maintenance of boundary cap neural crest stem cells in vitro. J Biomed Mater Res A 2020; 108:1274-1280. [PMID: 32061005 DOI: 10.1002/jbm.a.36900] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2019] [Revised: 02/08/2020] [Accepted: 02/10/2020] [Indexed: 12/20/2022]
Abstract
Optimal combination of stem cells and biocompatible support material is a promising strategy for successful tissue engineering. The required differentiation of stem cells is crucial for functionality of engineered tissues and can be regulated by chemical and physical cues. Here we examined how boundary cap neural crest stem cells (bNCSCs) are affected when cultured in the same medium, but on collagen- or laminin-polyacrylamide (PAA) scaffolds of different stiffness (0.5, 1, or ~7 kPa). bNCSCs displayed marked differences in their ability to attach, maintain a large cell population and differentiate, depending on scaffold stiffness. These findings show that the design of physical cues is an important parameter to achieve optimal stem cell properties for tissue repair and engineering.
Collapse
Affiliation(s)
- Yilin Han
- Department of Neuroscience, Uppsala University, Biomedical Centre, Uppsala, Sweden
| | - Daiva Baltriukienė
- Institute of Biochemistry, Life Sciences Center, Vilnius University, Vilnius, Lithuania
| | - Elena N Kozlova
- Department of Neuroscience, Uppsala University, Biomedical Centre, Uppsala, Sweden
| |
Collapse
|
30
|
Stierli S, Imperatore V, Lloyd AC. Schwann cell plasticity-roles in tissue homeostasis, regeneration, and disease. Glia 2019; 67:2203-2215. [PMID: 31215712 DOI: 10.1002/glia.23643] [Citation(s) in RCA: 80] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2019] [Revised: 05/14/2019] [Accepted: 05/14/2019] [Indexed: 12/12/2022]
Abstract
How tissues are maintained over a lifetime and repaired following injury are fundamental questions in biology with a disruption to these processes underlying pathologies such as cancer and degenerative disorders. It is becoming increasingly clear that each tissue has a distinct mechanism to maintain homeostasis and respond to injury utilizing different types of stem/progenitor cell populations depending on the insult and/or with a contribution from more differentiated cells that are able to dedifferentiate to aid tissue regeneration. Peripheral nerves are highly quiescent yet show remarkable regenerative capabilities. Remarkably, there is no evidence for a classical stem cell population, rather all cell-types within the nerve are able to proliferate to produce new nerve tissue. Co-ordinating the regeneration of this tissue are Schwann cells (SCs), the main glial cells of the peripheral nervous system. SCs exist in architecturally stable structures that can persist for the lifetime of an animal, however, they are not postmitotic, in that following injury they are reprogrammed at high efficiency to a progenitor-like state, with these cells acting to orchestrate the nerve regeneration process. During nerve regeneration, SCs show little plasticity, maintaining their identity in the repaired tissue. However, once free of the nerve environment they appear to exhibit increased plasticity with reported roles in the repair of other tissues. In this review, we will discuss the mechanisms underlying the homeostasis and regeneration of peripheral nerves and how reprogrammed progenitor-like SCs have broader roles in the repair of other tissues with implications for pathologies such as cancer.
Collapse
Affiliation(s)
- Salome Stierli
- MRC LMCB, University College London, Gower Street, London, WC1E 6BT, UK
| | | | - Alison C Lloyd
- MRC LMCB, University College London, Gower Street, London, WC1E 6BT, UK
| |
Collapse
|
31
|
The evolution and multi-molecular properties of NF1 cutaneous neurofibromas originating from C-fiber sensory endings and terminal Schwann cells at normal sites of sensory terminations in the skin. PLoS One 2019; 14:e0216527. [PMID: 31107888 PMCID: PMC6527217 DOI: 10.1371/journal.pone.0216527] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2018] [Accepted: 04/24/2019] [Indexed: 12/30/2022] Open
Abstract
In addition to large plexiform neurofibromas (pNF), NF1 patients are frequently disfigured by cutaneous neurofibromas (cNF) and are often afflicted with chronic pain and itch even from seemingly normal skin areas. Both pNFs and cNF consist primarily of benign hyperproliferating nonmyelinating Schwann cells (nSC). While pNF clearly arise within deep nerves and plexuses, the role of cutaneous innervation in the origin of cNF and in chronic itch and pain is unknown. First, we conducted a comprehensive, multi-molecular, immunofluorescence (IF) analyses on 3mm punch biopsies from three separate locations in normal appearing, cNF-free skin in 19 NF1 patients and skin of 16 normal subjects. At least one biopsy in 17 NF1 patients had previously undescribed micro-lesions consisting of a small, dense cluster of nonpeptidergic C-fiber endings and the affiliated nSC consistently adjoining adnexal structures—dermal papillae, hair follicles, sweat glands, sweat ducts, and arterioles—where C-fiber endings normally terminate. Similar micro-lesions were detected in hind paw skin of mice with conditionally-induced SC Nf1-/- mutations. Hypothesizing that these microlesions were pre-cNF origins of cNF, we subsequently analyzed numerous overt, small cNF (s-cNF, 3–6 mm) and discovered that each had an adnexal structure at the epicenter of vastly increased nonpeptidergic C-fiber terminals, accompanied by excessive nSC. The IF and functional genomics assays indicated that neurturin (NTRN) and artemin (ARTN) signaling through cRET kinase and GFRα2 and GFRα3 co-receptors on the aberrant C-fiber endings and nSC may mutually promote the onset of pre-cNF and their evolution to s-cNF. Moreover, TrpA1 and TrpV1 receptors may, respectively, mediate symptoms of chronic itch and pain. These newly discovered molecular characteristics might be targeted to suppress the development of cNF and to treat chronic itch and pain symptoms in NF1 patients.
Collapse
|
32
|
Jessen KR, Mirsky R. Schwann Cell Precursors; Multipotent Glial Cells in Embryonic Nerves. Front Mol Neurosci 2019; 12:69. [PMID: 30971890 PMCID: PMC6443887 DOI: 10.3389/fnmol.2019.00069] [Citation(s) in RCA: 117] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2018] [Accepted: 03/04/2019] [Indexed: 12/20/2022] Open
Abstract
The cells of the neural crest, often referred to as neural crest stem cells, give rise to a number of sub-lineages, one of which is Schwann cells, the glial cells of peripheral nerves. Crest cells transform to adult Schwann cells through the generation of two well defined intermediate stages, the Schwann cell precursors (SCP) in early embryonic nerves, and immature Schwann cells (iSch) in late embryonic and perinatal nerves. SCP are formed when neural crest cells enter nascent nerves and form intimate relationships with axons, a diagnostic feature of glial cells. This involves large-scale changes in gene expression, including the activation of established glial cell markers. Like early glia in the CNS, radial glia, SCP retain developmental multipotency and contribute to other crest-derived lineages during embryonic development. SCP, as well as closely related cells termed boundary cap cells, and later stages of the Schwann cell lineage have all been implicated as the tumor initiating cell in NF1 associated neurofibromas. iSch are formed from SCP in a process that involves the appearance of additional differentiation markers, autocrine survival circuits, cellular elongation, a formation of endoneurial connective tissue and basal lamina. Finally, in peri- and post-natal nerves, iSch are reversibly induced by axon-associated signals to form the myelin and non-myelin Schwann cells of adult nerves. This review article discusses early Schwann cell development in detail and describes a large number of molecular signaling systems that control glial development in embryonic nerves.
Collapse
Affiliation(s)
- Kristjan R. Jessen
- Department of Cell and Developmental Biology, University College London, London, United Kingdom
| | | |
Collapse
|
33
|
Morikawa S, Iribar H, Gutiérrez-Rivera A, Ezaki T, Izeta A. Pericytes in Cutaneous Wound Healing. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1147:1-63. [DOI: 10.1007/978-3-030-16908-4_1] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
34
|
Injury and stress responses of adult neural crest-derived cells. Dev Biol 2018; 444 Suppl 1:S356-S365. [DOI: 10.1016/j.ydbio.2018.05.011] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Revised: 05/15/2018] [Accepted: 05/15/2018] [Indexed: 12/21/2022]
|
35
|
Fontenas L, Kucenas S. Motor Exit Point (MEP) Glia: Novel Myelinating Glia That Bridge CNS and PNS Myelin. Front Cell Neurosci 2018; 12:333. [PMID: 30356886 PMCID: PMC6190867 DOI: 10.3389/fncel.2018.00333] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2018] [Accepted: 09/11/2018] [Indexed: 12/12/2022] Open
Abstract
Oligodendrocytes (OLs) and Schwann cells (SCs) have traditionally been thought of as the exclusive myelinating glial cells of the central and peripheral nervous systems (CNS and PNS), respectively, for a little over a century. However, recent studies demonstrate the existence of a novel, centrally-derived peripheral glial population called motor exit point (MEP) glia, which myelinate spinal motor root axons in the periphery. Until recently, the boundaries that exist between the CNS and PNS, and the cells permitted to cross them, were mostly described based on fixed histological collections and static lineage tracing. Recent work in zebrafish using in vivo, time-lapse imaging has shed light on glial cell interactions at the MEP transition zone and reveals a more complex picture of myelination both centrally and peripherally.
Collapse
Affiliation(s)
- Laura Fontenas
- Department of Biology, University of Virginia, Charlottesville, VA, United States
| | - Sarah Kucenas
- Department of Biology, University of Virginia, Charlottesville, VA, United States
| |
Collapse
|
36
|
Angelim MKSC, Maia LMSDS, Mouffle C, Ginhoux F, Low D, Amancio-Dos-Santos A, Makhoul J, Le Corronc H, Mangin JM, Legendre P. Embryonic macrophages and microglia ablation alter the development of dorsal root ganglion sensory neurons in mouse embryos. Glia 2018; 66:2470-2486. [PMID: 30252950 DOI: 10.1002/glia.23499] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Revised: 06/29/2018] [Accepted: 07/03/2018] [Indexed: 12/15/2022]
Abstract
Microglia are known to regulate several aspects of the development of the central nervous system. When microglia colonize the spinal cord, from E11.5 in the mouse embryo, they interact with growing central axons of dorsal root ganglion sensory neurons (SNs), which suggests that they may have some functions in SN development. To address this issue, we analyzed the effects of embryonic macrophage ablation on the early development of SNs using mouse embryo lacking embryonic macrophages (PU.1 knock-out mice) and immune cell ablation. We discovered that, in addition to microglia, embryonic macrophages contact tropomyosin receptor kinase (Trk) C+ SN, TrkB+ SN, and TrkA+ SN peripheral neurites from E11.5. Deprivation of immune cells resulted in an initial reduction of TrkC+ SN and TrkB+ SN populations at E11.5 that was unlikely to be related to an alteration in their developmental cell death (DCD), followed by a transitory increase in their number at E12.5. It also resulted in a reduction of TrkA+ SN number during the developmental period analyzed (E11.5-E15.5), although we did not observe any change in their DCD. Proliferation of cells negative for brain fatty acid-binding protein (BFABP- ), which likely correspond to neuronal progenitors, was increased at E11.5, while their proliferation was decreased at E12.5, which could partly explain the alterations of SN subtype production observed from E11.5. In addition, we observed alterations in the proliferation of glial cell progenitors (BFABP+ cells) in the absence of embryonic macrophages. Our data indicate that embryonic macrophages and microglia ablation alter the development of SNs.
Collapse
Affiliation(s)
- Monara Kaélle Sérvulo Cruz Angelim
- Sorbonne Université, UPMC Univ Paris 06, INSERM, CNRS, Neurosciences Paris Seine, Institut de Biologie Paris Seine (NPS, IBPS), Paris, France.,Neurophysiology and pharmacology laboratory, Federal University of Pernambuco, Pernambuco, Brazil
| | - Luciana Maria Silva de Seixas Maia
- Sorbonne Université, UPMC Univ Paris 06, INSERM, CNRS, Neurosciences Paris Seine, Institut de Biologie Paris Seine (NPS, IBPS), Paris, France.,Neurophysiology and pharmacology laboratory, Federal University of Pernambuco, Pernambuco, Brazil
| | - Christine Mouffle
- Sorbonne Université, UPMC Univ Paris 06, INSERM, CNRS, Neurosciences Paris Seine, Institut de Biologie Paris Seine (NPS, IBPS), Paris, France
| | - Florent Ginhoux
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research, Singapore, Singapore
| | - Donovan Low
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research, Singapore, Singapore
| | | | - Jennifer Makhoul
- Sorbonne Université, UPMC Univ Paris 06, INSERM, CNRS, Neurosciences Paris Seine, Institut de Biologie Paris Seine (NPS, IBPS), Paris, France
| | - Hervé Le Corronc
- Sorbonne Université, UPMC Univ Paris 06, INSERM, CNRS, Neurosciences Paris Seine, Institut de Biologie Paris Seine (NPS, IBPS), Paris, France.,Université d'Angers, Angers, France
| | - Jean-Marie Mangin
- Sorbonne Université, UPMC Univ Paris 06, INSERM, CNRS, Neurosciences Paris Seine, Institut de Biologie Paris Seine (NPS, IBPS), Paris, France
| | - Pascal Legendre
- Sorbonne Université, UPMC Univ Paris 06, INSERM, CNRS, Neurosciences Paris Seine, Institut de Biologie Paris Seine (NPS, IBPS), Paris, France
| |
Collapse
|
37
|
Furlan A, Adameyko I. Schwann cell precursor: a neural crest cell in disguise? Dev Biol 2018; 444 Suppl 1:S25-S35. [PMID: 29454705 DOI: 10.1016/j.ydbio.2018.02.008] [Citation(s) in RCA: 104] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2017] [Revised: 02/14/2018] [Accepted: 02/14/2018] [Indexed: 01/19/2023]
Abstract
Schwann cell precursors (SCPs) are multipotent embryonic progenitors covering all developing peripheral nerves. These nerves grow and navigate with unprecedented precision, delivering SCP progenitors to almost all locations in the embryonic body. Within specific developing tissues, SCPs detach from nerves and generate neuroendocrine cells, autonomic neurons, mature Schwann cells, melanocytes and other cell types. These properties of SCPs evoke resemblances between them and their parental population, namely, neural crest cells. Neural crest cells are incredibly multipotent migratory cells that revolutionized the course of evolution in the lineage of early chordate animals. Given this similarity and recent data, it is possible to hypothesize that proto-neural crest cells are similar to SCPs spreading along the nerves. Here, we review the multipotency of SCPs, the signals that govern them, their potential therapeutic value, SCP's embryonic origin and their evolutionary connections. We dedicate this article to the memory of Wilhelm His, the father of the microtome and "Zwischenstrang", currently known as the neural crest.
Collapse
Affiliation(s)
- Alessandro Furlan
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York 11724 USA
| | - Igor Adameyko
- Department of Physiology and Pharmacology, Karolinska Institutet, 17177 Stockholm, Sweden; Center for Brain Research, Medical University Vienna, 1090 Vienna, Austria.
| |
Collapse
|