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
|
Kolos EA, Korzhevskii DE. Ventral Root Boundary Cap Cells of Rat Spinal Cord Contain Connexin-43. Bull Exp Biol Med 2024:10.1007/s10517-024-06299-2. [PMID: 39589621 DOI: 10.1007/s10517-024-06299-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Indexed: 11/27/2024]
Abstract
Boundary cap cells are a population of multipotent stem cells that have great potential for the use in the treatment of damaged nervous system. We studied the patterns of distribution of the gap junction protein connexin-43 (Cx43) in boundary cap cells of the ventral root of the spinal cord of rat embryos (E12-E20; n=40). It was found that Cx43 is expressed in ventral boundary cap cells at all stages of its existence during embryogenesis. At the early stages of prenatal development, the cytoplasmic distribution of Cx43 in the boundary cap cells predominates; at the later stages, Cx43-immunopositive punctate structures are identified. These puncta represent gap junction plaques between the cells. It can be assumed that during the early embryogenesis, Cx43 regulates the main histogenetic processes in boundary cap cells and only in the later stages of prenatal development, Cx43-mediated communications are formed between boundary cap cells.
Collapse
Affiliation(s)
- E A Kolos
- Institute of Experimental Medicine, St. Petersburg, Russia.
| | | |
Collapse
|
3
|
Erickson AG, Motta A, Kastriti ME, Edwards S, Coulpier F, Théoulle E, Murtazina A, Poverennaya I, Wies D, Ganofsky J, Canu G, Lallemend F, Topilko P, Hadjab S, Fried K, Ruhrberg C, Schwarz Q, Castellani V, Bonanomi D, Adameyko I. Motor innervation directs the correct development of the mouse sympathetic nervous system. Nat Commun 2024; 15:7065. [PMID: 39152112 PMCID: PMC11329663 DOI: 10.1038/s41467-024-51290-0] [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: 06/09/2021] [Accepted: 08/02/2024] [Indexed: 08/19/2024] Open
Abstract
The sympathetic nervous system controls bodily functions including vascular tone, cardiac rhythm, and the "fight-or-flight response". Sympathetic chain ganglia develop in parallel with preganglionic motor nerves extending from the neural tube, raising the question of whether axon targeting contributes to sympathetic chain formation. Using nerve-selective genetic ablations and lineage tracing in mouse, we reveal that motor nerve-associated Schwann cell precursors (SCPs) contribute sympathetic neurons and satellite glia after the initial seeding of sympathetic ganglia by neural crest. Motor nerve ablation causes mispositioning of SCP-derived sympathoblasts as well as sympathetic chain hypoplasia and fragmentation. Sympathetic neurons in motor-ablated embryos project precociously and abnormally towards dorsal root ganglia, eventually resulting in fusion of sympathetic and sensory ganglia. Cell interaction analysis identifies semaphorins as potential motor nerve-derived signaling molecules regulating sympathoblast positioning and outgrowth. Overall, central innervation functions both as infrastructure and regulatory niche to ensure the integrity of peripheral ganglia morphogenesis.
Collapse
Affiliation(s)
- Alek G Erickson
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Alessia Motta
- Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milano, Italy
| | - Maria Eleni Kastriti
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
- Center for Brain Research, Department of Neuroimmunology, Medical University Vienna, Vienna, Austria
| | - Steven Edwards
- Department of Applied Physics, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Fanny Coulpier
- Mondor Institute for Biomedical Research (IMRB), INSERM, Créteil, France
| | - Emy Théoulle
- University of Claude Bernard Lyon 1, MeLiS, CNRS, INSERM, NeuroMyoGene Institute, Lyon, France
| | - Aliia Murtazina
- Department of Neuroscience, Biomedicum, Karolinska Institute, Stockholm, Sweden
| | - Irina Poverennaya
- Center for Brain Research, Department of Neuroimmunology, Medical University Vienna, Vienna, Austria
| | - Daniel Wies
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Jeremy Ganofsky
- University of Claude Bernard Lyon 1, MeLiS, CNRS, INSERM, NeuroMyoGene Institute, Lyon, France
| | - Giovanni Canu
- University College London, Department of Ophthalmology London, London, UK
| | - Francois Lallemend
- Department of Neuroscience, Biomedicum, Karolinska Institute, Stockholm, Sweden
| | - Piotr Topilko
- Mondor Institute for Biomedical Research (IMRB), INSERM, Créteil, France
| | - Saida Hadjab
- Department of Neuroscience, Biomedicum, Karolinska Institute, Stockholm, Sweden
| | - Kaj Fried
- Department of Neuroscience, Biomedicum, Karolinska Institute, Stockholm, Sweden
| | | | - Quenten Schwarz
- Center for Cancer Biology, University of South Australia, Adelaide, SA, Australia
| | - Valerie Castellani
- University of Claude Bernard Lyon 1, MeLiS, CNRS, INSERM, NeuroMyoGene Institute, Lyon, France
| | - Dario Bonanomi
- Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milano, Italy.
| | - Igor Adameyko
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden.
- Center for Brain Research, Department of Neuroimmunology, Medical University Vienna, Vienna, Austria.
| |
Collapse
|
4
|
Perrin S, Protic S, Bretegnier V, Laurendeau I, de Lageneste OD, Panara N, Ruckebusch O, Luka M, Masson C, Maillard T, Coulpier F, Pannier S, Wicart P, Hadj-Rabia S, Radomska KJ, Zarhrate M, Ménager M, Vidaud D, Topilko P, Parfait B, Colnot C. MEK-SHP2 inhibition prevents tibial pseudarthrosis caused by NF1 loss in Schwann cells and skeletal stem/progenitor cells. Sci Transl Med 2024; 16:eadj1597. [PMID: 38924432 DOI: 10.1126/scitranslmed.adj1597] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Revised: 01/15/2024] [Accepted: 05/24/2024] [Indexed: 06/28/2024]
Abstract
Congenital pseudarthrosis of the tibia (CPT) is a severe pathology marked by spontaneous bone fractures that fail to heal, leading to fibrous nonunion. Half of patients with CPT are affected by the multisystemic genetic disorder neurofibromatosis type 1 (NF1) caused by mutations in the NF1 tumor suppressor gene, a negative regulator of RAS-mitogen-activated protein kinase (MAPK) signaling pathway. Here, we analyzed patients with CPT and Prss56-Nf1 knockout mice to elucidate the pathogenic mechanisms of CPT-related fibrous nonunion and explored a pharmacological approach to treat CPT. We identified NF1-deficient Schwann cells and skeletal stem/progenitor cells (SSPCs) in pathological periosteum as affected cell types driving fibrosis. Whereas NF1-deficient SSPCs adopted a fibrotic fate, NF1-deficient Schwann cells produced critical paracrine factors including transforming growth factor-β and induced fibrotic differentiation of wild-type SSPCs. To counteract the elevated RAS-MAPK signaling in both NF1-deficient Schwann cells and SSPCs, we used MAPK kinase (MEK) and Src homology 2 containing protein tyrosine phosphatase 2 (SHP2) inhibitors. Combined MEK-SHP2 inhibition in vivo prevented fibrous nonunion in the Prss56-Nf1 knockout mouse model, providing a promising therapeutic strategy for the treatment of fibrous nonunion in CPT.
Collapse
Affiliation(s)
- Simon Perrin
- Université Paris Est Creteil, INSERM, IMRB, 94000 Creteil, France
| | - Sanela Protic
- Université Paris Est Creteil, INSERM, IMRB, 94000 Creteil, France
| | | | - Ingrid Laurendeau
- INSERM UMR S1016, Institut Cochin, Université Paris Cité, 75014 Paris, France
| | | | - Nicolas Panara
- INSERM UMR S1016, Institut Cochin, Université Paris Cité, 75014 Paris, France
| | - Odile Ruckebusch
- Université Paris Est Creteil, INSERM, IMRB, Plateforme de Cytométrie en flux, 94000 Creteil, France
| | - Marine Luka
- Paris Cité University, Imagine Institute, Laboratory of Inflammatory Responses and Transcriptomic Networks in Diseases, Atip-Avenir Team, INSERM UMR 1163, 75015 Paris, France
- Labtech Single-Cell@Imagine, Imagine Institute, INSERM UMR 1163, 75015 Paris, France
| | - Cécile Masson
- Bioinformatics Core Facility, Institut Imagine-Structure Fédérative de Recherche Necker, INSERM U1163, 75015 Paris, France
- INSERM US24/CNRS UAR3633, Paris Cité University, 75015 Paris, France
| | - Théodora Maillard
- Service de Médecine Génomique des Maladies de Système et d'Organe, Hôpital Cochin, DMU BioPhyGen, Assistance Publique-Hôpitaux de Paris, AP-HP, Centre-Université Paris Cité, F-75014 Paris, France
| | - Fanny Coulpier
- Université Paris Est Creteil, INSERM, IMRB, 94000 Creteil, France
| | - Stéphanie Pannier
- Department of Pediatric Orthopedic Surgery and Traumatology, Necker-Enfants Malades Hospital, AP-HP, Paris Cité University, 75015 Paris, France
| | - Philippe Wicart
- Department of Pediatric Orthopedic Surgery and Traumatology, Necker-Enfants Malades Hospital, AP-HP, Paris Cité University, 75015 Paris, France
| | - Smail Hadj-Rabia
- Department of Dermatology, Reference Center for Rare Skin Diseases (MAGEC), Imagine Institute, Necker-Enfants Malades Hospital, AP-HP, Paris Cité University, 75015 Paris, France
| | | | - Mohammed Zarhrate
- INSERM US24/CNRS UAR3633, Paris Cité University, 75015 Paris, France
- Genomics Core Facility, Institut Imagine-Structure Fédérative de Recherche Necker, INSERM U1163, 75015 Paris, France
| | - Mickael Ménager
- Paris Cité University, Imagine Institute, Laboratory of Inflammatory Responses and Transcriptomic Networks in Diseases, Atip-Avenir Team, INSERM UMR 1163, 75015 Paris, France
- Labtech Single-Cell@Imagine, Imagine Institute, INSERM UMR 1163, 75015 Paris, France
| | - Dominique Vidaud
- INSERM UMR S1016, Institut Cochin, Université Paris Cité, 75014 Paris, France
- Service de Médecine Génomique des Maladies de Système et d'Organe, Hôpital Cochin, DMU BioPhyGen, Assistance Publique-Hôpitaux de Paris, AP-HP, Centre-Université Paris Cité, F-75014 Paris, France
| | - Piotr Topilko
- Université Paris Est Creteil, INSERM, IMRB, 94000 Creteil, France
| | - Béatrice Parfait
- INSERM UMR S1016, Institut Cochin, Université Paris Cité, 75014 Paris, France
- Service de Médecine Génomique des Maladies de Système et d'Organe, Hôpital Cochin, DMU BioPhyGen, Assistance Publique-Hôpitaux de Paris, AP-HP, Centre-Université Paris Cité, F-75014 Paris, France
| | - Céline Colnot
- Université Paris Est Creteil, INSERM, IMRB, 94000 Creteil, France
| |
Collapse
|
5
|
Hastings RL, Valdez G. Origin, identity, and function of terminal Schwann cells. Trends Neurosci 2024; 47:432-446. [PMID: 38664109 PMCID: PMC11168889 DOI: 10.1016/j.tins.2024.03.007] [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: 11/28/2023] [Revised: 02/20/2024] [Accepted: 03/26/2024] [Indexed: 06/14/2024]
Abstract
The highly specialized nonmyelinating glial cells present at somatic peripheral nerve endings, known collectively as terminal Schwann cells (TSCs), play critical roles in the development, function and repair of their motor and sensory axon terminals and innervating tissue. Over the past decades, research efforts across various vertebrate species have revealed that while TSCs are a diverse group of cells, they share a number of features among them. In this review, we summarize the state-of-knowledge about each TSC type and explore the opportunities that TSCs provide to treat conditions that afflict peripheral axon terminals.
Collapse
Affiliation(s)
- Robert Louis Hastings
- Department of Molecular Biology, Cell Biology and Biochemistry, Brown University, Providence, RI, USA
| | - Gregorio Valdez
- Department of Molecular Biology, Cell Biology and Biochemistry, Brown University, Providence, RI, USA; Center for Translational Neuroscience, Robert J. and Nancy D. Carney Institute for Brain Science, and Center on the Biology of Aging, Brown University, Providence, RI, USA.
| |
Collapse
|
6
|
Trolle C, Han Y, Mutt SJ, Christoffersson G, Kozlova EN. Boundary cap neural crest stem cells promote angiogenesis after transplantation to avulsed dorsal roots in mice and induce migration of endothelial cells in 3D printed scaffolds. Neurosci Lett 2024; 826:137724. [PMID: 38467271 DOI: 10.1016/j.neulet.2024.137724] [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: 11/25/2023] [Revised: 02/26/2024] [Accepted: 03/08/2024] [Indexed: 03/13/2024]
Abstract
Dorsal root avulsion injuries lead to loss of sensation and to reorganization of blood vessels (BVs) in the injured area. The inability of injured sensory axons to re-enter the spinal cord results in permanent loss of sensation, and often also leads to the development of neuropathic pain. Approaches that restore connection between peripheral sensory axons and their CNS targets are thus urgently need. Previous research has shown that sensory axons from peripherally grafted human sensory neurons are able to enter the spinal cord by growing along BVs which penetrate the CNS from the spinal cord surface. In this study we analysed the distribution of BVs after avulsion injury and how their pattern is affected by implantation at the injury site of boundary cap neural crest stem cells (bNCSCs), a transient cluster of cells, which are located at the boundary between the spinal cord and peripheral nervous system and assist the growth of sensory axons from periphery into the spinal cord during development. The superficial dorsal spinal cord vasculature was examined using intravital microscopy and intravascular BV labelling. bNCSC transplantation increased vascular volume in a non-dose responsive manner, whereas dorsal root avulsion alone did not decrease the vascular volume. To determine whether bNCSC are endowed with angiogenic properties we prepared 3D printed scaffolds, containing bNCSCs together with rings prepared from mouse aorta. We show that bNCSC do induce migration and assembly of endothelial cells in this system. These findings suggest that bNCSC transplant can promote vascularization in vivo and contribute to BV formation in 3D printed scaffolds.
Collapse
Affiliation(s)
- Carl Trolle
- Department of Medical Sciences, Uppsala University Hospital, Rehabilitation Medicine, 751 85 Uppsala, Sweden
| | - Yilin Han
- Department of Immunology, Genetics and Pathology, Uppsala University Biomedical Center, PO Box 815, 751 08 Uppsala, Sweden
| | - Shivaprakash Jagalur Mutt
- Department of Medical Cell Biology, Science for Life Laboratory, Uppsala University, PO Box 571, 751 23 Uppsala, Sweden
| | - Gustaf Christoffersson
- Department of Medical Cell Biology, Science for Life Laboratory, Uppsala University, PO Box 571, 751 23 Uppsala, Sweden
| | - Elena N Kozlova
- Department of Immunology, Genetics and Pathology, Uppsala University Biomedical Center, PO Box 815, 751 08 Uppsala, Sweden.
| |
Collapse
|
7
|
Gerschenfeld G, Coulpier F, Gresset A, Pulh P, Job B, Topilko T, Siegenthaler J, Kastriti ME, Brunet I, Charnay P, Topilko P. Neural tube-associated boundary caps are a major source of mural cells in the skin. eLife 2023; 12:e69413. [PMID: 38095361 PMCID: PMC10786459 DOI: 10.7554/elife.69413] [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: 04/14/2021] [Accepted: 12/12/2023] [Indexed: 01/13/2024] Open
Abstract
In addition to their roles in protecting nerves and increasing conduction velocity, peripheral glia plays key functions in blood vessel development by secreting molecules governing arteries alignment and maturation with nerves. Here, we show in mice that a specific, nerve-attached cell population, derived from boundary caps (BCs), constitutes a major source of mural cells for the developing skin vasculature. Using Cre-based reporter cell tracing and single-cell transcriptomics, we show that BC derivatives migrate into the skin along the nerves, detach from them, and differentiate into pericytes and vascular smooth muscle cells. Genetic ablation of this population affects the organization of the skin vascular network. Our results reveal the heterogeneity and extended potential of the BC population in mice, which gives rise to mural cells, in addition to previously described neurons, Schwann cells, and melanocytes. Finally, our results suggest that mural specification of BC derivatives takes place before their migration along nerves to the mouse skin.
Collapse
Affiliation(s)
- Gaspard Gerschenfeld
- Institut de Biologie de l'Ecole normale supérieure (IBENS), Ecole normale supérieure, CNRS, Inserm, Université PSLParisFrance
- Sorbonne Université, Collège DoctoralParisFrance
| | - Fanny Coulpier
- Institut de Biologie de l'Ecole normale supérieure (IBENS), Ecole normale supérieure, CNRS, Inserm, Université PSLParisFrance
- nstitut Mondor de Recherche Biomédicale, Inserm U955-Team 9CréteilFrance
- Genomic facility, Ecole normale supérieure, PSL Research University, CNRS, Inserm, Institut de Biologie de l'Ecole normale supérieure (IBENS)ParisFrance
| | - Aurélie Gresset
- Institut de Biologie de l'Ecole normale supérieure (IBENS), Ecole normale supérieure, CNRS, Inserm, Université PSLParisFrance
| | - Pernelle Pulh
- Institut de Biologie de l'Ecole normale supérieure (IBENS), Ecole normale supérieure, CNRS, Inserm, Université PSLParisFrance
- nstitut Mondor de Recherche Biomédicale, Inserm U955-Team 9CréteilFrance
| | - Bastien Job
- Inserm US23, AMMICA, Institut Gustave RoussyVillejuifFrance
| | - Thomas Topilko
- Laboratoire de Plasticité Structurale, Sorbonne Université, ICM Institut du Cerveau et de la Moelle Epinière, Inserm U1127, CNRS UMR7225ParisFrance
| | - Julie Siegenthaler
- Department of Pediatrics Section of Developmental Biology, University of Colorado Anschutz Medical CampusAuroraUnited States
| | - Maria Eleni Kastriti
- Department of Physiology and Pharmacology, Karolinska InstitutetStockholmSweden
- Department of Neuroimmunology, Center for Brain Research, Medical University ViennaViennaAustria
| | - Isabelle Brunet
- Inserm U1050, Centre Interdisciplinaire de Recherche en Biologie (CIRB), Collège de FranceParisFrance
| | - Patrick Charnay
- Institut de Biologie de l'Ecole normale supérieure (IBENS), Ecole normale supérieure, CNRS, Inserm, Université PSLParisFrance
| | - Piotr Topilko
- Institut de Biologie de l'Ecole normale supérieure (IBENS), Ecole normale supérieure, CNRS, Inserm, Université PSLParisFrance
- nstitut Mondor de Recherche Biomédicale, Inserm U955-Team 9CréteilFrance
| |
Collapse
|
8
|
Coulpier F, Pulh P, Oubrou L, Naudet J, Fertitta L, Gregoire JM, Bocquet A, Schmitt AM, Wolkenstein P, Radomska KJ, Topilko P. Topical delivery of mitogen-activated protein kinase inhibitor binimetinib prevents the development of cutaneous neurofibromas in neurofibromatosis type 1 mutant mice. Transl Res 2023; 261:16-27. [PMID: 37331503 DOI: 10.1016/j.trsl.2023.06.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Revised: 05/19/2023] [Accepted: 06/13/2023] [Indexed: 06/20/2023]
Abstract
Cutaneous neurofibromas (cNFs) are a hallmark of patients with the neurofibromatosis type 1 (NF1) genetic disorder. These benign nerve sheath tumors, which can amount to thousands, develop from puberty onward, often cause pain and are considered by patients to be the primary burden of the disease. Mutations of NF1, encoding a negative regulator of the RAS signaling pathway, in the Schwann cell (SCs) lineage are considered to be at the origin of cNFs. The mechanisms governing cNFs development are poorly understood, and therapeutics to reduce cNFs are missing, mainly due to the lack of appropriate animal models. To address this, we designed the Nf1-KO mouse model that develops cNFs. Using this model, we found that cNFs development is a singular event and goes through 3 successive stages: initiation, progression, and stabilization characterized by changes in the proliferative and MAPK activities of tumor SCs. We found that skin trauma accelerated the development of cNFs and further used this model to explore the efficacy of the MEK inhibitor binimetinib to cure these tumors. We showed that while topically delivered binimetinib has a selective and minor effect on mature cNFs, the same drug prevents their development over long periods.
Collapse
Affiliation(s)
- Fanny Coulpier
- Mondor Institute for Biomedical Research, Creteil, France
| | - Pernelle Pulh
- Mondor Institute for Biomedical Research, Creteil, France
| | - Layna Oubrou
- Mondor Institute for Biomedical Research, Creteil, France
| | - Julie Naudet
- Mondor Institute for Biomedical Research, Creteil, France
| | - Laura Fertitta
- Mondor Institute for Biomedical Research, Creteil, France; Dermatology Department, Centre de Référence des Neurofibromatoses, Hôpital Henri-Mondor, AP-HP, Créteil, France
| | | | | | | | - Pierre Wolkenstein
- Mondor Institute for Biomedical Research, Creteil, France; Dermatology Department, Centre de Référence des Neurofibromatoses, Hôpital Henri-Mondor, AP-HP, Créteil, France
| | | | - Piotr Topilko
- Mondor Institute for Biomedical Research, Creteil, France.
| |
Collapse
|
9
|
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
|
10
|
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
|
11
|
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
|
12
|
Marechal E, Poliard A, Henry K, Moreno M, Legrix M, Macagno N, Mondielli G, Fauquier T, Barlier A, Etchevers HC. Multiple congenital malformations arise from somatic mosaicism for constitutively active Pik3ca signaling. Front Cell Dev Biol 2022; 10:1013001. [PMID: 36353506 PMCID: PMC9637999 DOI: 10.3389/fcell.2022.1013001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2022] [Accepted: 10/07/2022] [Indexed: 11/17/2022] Open
Abstract
Recurrent missense mutations of the PIK3CA oncogene are among the most frequent drivers of human cancers. These often lead to constitutive activation of its product p110α, a phosphatidylinositol 3-kinase (PI3K) catalytic subunit. In addition to causing a broad range of cancers, the H1047R mutation is also found in affected tissues of a distinct set of congenital tumors and malformations. Collectively termed PIK3CA-related disorders (PRDs), these lead to overgrowth of brain, adipose, connective and musculoskeletal tissues and/or blood and lymphatic vessel components. Vascular malformations are frequently observed in PRD, due to cell-autonomous activation of PI3K signaling within endothelial cells. These, like most muscle, connective tissue and bone, are derived from the embryonic mesoderm. However, important organ systems affected in PRDs are neuroectodermal derivatives. To further examine their development, we drove the most common post-zygotic activating mutation of Pik3ca in neural crest and related embryonic lineages. Outcomes included macrocephaly, cleft secondary palate and more subtle skull anomalies. Surprisingly, Pik3ca-mutant subpopulations of neural crest origin were also associated with widespread cephalic vascular anomalies. Mesectodermal neural crest is a major source of non-endothelial connective tissue in the head, but not the body. To examine the response of vascular connective tissues of the body to constitutive Pik3ca activity during development, we expressed the mutation by way of an Egr2 (Krox20) Cre driver. Lineage tracing led us to observe new lineages that had normally once expressed Krox20 and that may be co-opted in pathogenesis, including vascular pericytes and perimysial fibroblasts. Finally, Schwann cell precursors having transcribed either Krox20 or Sox10 and induced to express constitutively active PI3K were associated with vascular and other tumors. These murine phenotypes may aid discovery of new candidate human PRDs affecting craniofacial and vascular smooth muscle development as well as the reciprocal paracrine signaling mechanisms leading to tissue overgrowth.
Collapse
Affiliation(s)
- Elise Marechal
- INSERM, MMG, U1251, MarMaRa Institute, Aix Marseille University, Marseille, France
| | - Anne Poliard
- URP 2496 Orofacial Pathologies, Imagery and Biotherapies, CNRS, GDR 2031 CREST-NET, Université Paris Cité, Montrouge, France
- School of Dentistry, Université Paris Cité, Montrouge, France
| | - Kilian Henry
- School of Dentistry, Université Paris Cité, Montrouge, France
| | - Mathias Moreno
- INSERM, MMG, U1251, MarMaRa Institute, Aix Marseille University, Marseille, France
| | - Mathilde Legrix
- INSERM, MMG, U1251, MarMaRa Institute, Aix Marseille University, Marseille, France
| | - Nicolas Macagno
- INSERM, MMG, U1251, MarMaRa Institute, Aix Marseille University, Marseille, France
| | - Grégoire Mondielli
- INSERM, MMG, U1251, MarMaRa Institute, Aix Marseille University, Marseille, France
| | - Teddy Fauquier
- INSERM, MMG, U1251, MarMaRa Institute, Aix Marseille University, Marseille, France
| | - Anne Barlier
- INSERM, MMG, U1251, MarMaRa Institute, Aix Marseille University, Marseille, France
- AP-HM, MMG, MarMaRa Institute, La Conception Hospital Laboratory of Molecular Biology, Marseille, France
| | - Heather C. Etchevers
- INSERM, MMG, U1251, CNRS, GDR 2031 CREST-NET, MarMaRa Institute, Aix Marseille University, Marseille, France
- *Correspondence: Heather C. Etchevers,
| |
Collapse
|
13
|
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
|
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
|
Suazo I, Vega JA, García-Mesa Y, García-Piqueras J, García-Suárez O, Cobo T. The Lamellar Cells of Vertebrate Meissner and Pacinian Corpuscles: Development, Characterization, and Functions. Front Neurosci 2022; 16:790130. [PMID: 35356056 PMCID: PMC8959428 DOI: 10.3389/fnins.2022.790130] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Accepted: 01/21/2022] [Indexed: 12/15/2022] Open
Abstract
Sensory corpuscles, or cutaneous end-organ complexes, are complex structures localized at the periphery of Aβ-axon terminals from primary sensory neurons that primarily work as low-threshold mechanoreceptors. Structurally, they consist, in addition to the axons, of non-myelinating Schwann-like cells (terminal glial cells) and endoneurial- and perineurial-related cells. The terminal glial cells are the so-called lamellar cells in Meissner and Pacinian corpuscles. Lamellar cells are variably arranged in sensory corpuscles as a “coin stack” in the Meissner corpuscles or as an “onion bulb” in the Pacinian ones. Nevertheless, the origin and protein profile of the lamellar cells in both morphotypes of sensory corpuscles is quite similar, although it differs in the expression of mechano-gated ion channels as well as in the composition of the extracellular matrix between the cells. The lamellar cells have been regarded as supportive cells playing a passive role in the process of genesis of the action potential, i.e., the mechanotransduction process. However, they express ion channels related to the mechano–electric transduction and show a synapse-like mechanism that suggest neurotransmission at the genesis of the electrical action potential. This review updates the current knowledge about the embryonic origin, development modifications, spatial arrangement, ultrastructural characteristics, and protein profile of the lamellar cells of cutaneous end-organ complexes focusing on Meissner and Pacinian morphotypes.
Collapse
Affiliation(s)
- Iván Suazo
- Grupo SINPOS, Departamento de Morfología y Biología Celular, Universidad de Oviedo, Oviedo, Spain
- Faculcultad de Ciencias de la Salud, Universidad Autónoma de Chile, Santiago, Chile
| | - José A. Vega
- Grupo SINPOS, Departamento de Morfología y Biología Celular, Universidad de Oviedo, Oviedo, Spain
- Faculcultad de Ciencias de la Salud, Universidad Autónoma de Chile, Santiago, Chile
- *Correspondence: José A. Vega,
| | - Yolanda García-Mesa
- Grupo SINPOS, Departamento de Morfología y Biología Celular, Universidad de Oviedo, Oviedo, Spain
| | - Jorge García-Piqueras
- Grupo SINPOS, Departamento de Morfología y Biología Celular, Universidad de Oviedo, Oviedo, Spain
| | - Olivia García-Suárez
- Grupo SINPOS, Departamento de Morfología y Biología Celular, Universidad de Oviedo, Oviedo, Spain
| | - Teresa Cobo
- Departamento de Cirugía y Especialidades Médico-Quirúrgicas, Universidad de Oviedo, Oviedo, Spain
| |
Collapse
|
17
|
Abstract
Neurofibromatosis type 1 (NF1) is one of the most common neurocutaneous genetic disorders, presenting with different cutaneous features such as café-au-lait macules, intertriginous skin freckling, and neurofibromas. Although most of the disease manifestations are benign, patients are at risk for a variety of malignancies, including malignant transformation of plexiform neurofibromas. Numerous studies have investigated the mechanisms by which these characteristic neurofibromas develop, with progress made toward unraveling the various players involved in their complex pathogenesis. In this review, we summarize the current understanding of the cells that give rise to NF1 neoplasms as well as the molecular mechanisms and cellular changes that confer tumorigenic potential. We also discuss the role of the tumor microenvironment and the key aspects of its various cell types that contribute to NF1-associated tumorigenesis. An increased understanding of these intrinsic and extrinsic components is critical for developing novel therapeutic approaches for affected patients.
Collapse
Affiliation(s)
- Ashley Bui
- Department of Pediatrics, The University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Chunhui Jiang
- Department of Dermatology, The University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Renee M McKay
- Department of Dermatology, The University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Laura J Klesse
- Department of Pediatrics, The University of Texas Southwestern Medical Center, Dallas, Texas, USA.,Harold C. Simmons Comprehensive Cancer Center, The University of Texas Southwestern Medical Center, Dallas, Texas, USA.,Comprehensive Neurofibromatosis Clinic, The University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Lu Q Le
- Department of Dermatology, The University of Texas Southwestern Medical Center, Dallas, Texas, USA.,Harold C. Simmons Comprehensive Cancer Center, The University of Texas Southwestern Medical Center, Dallas, Texas, USA.,Comprehensive Neurofibromatosis Clinic, The University of Texas Southwestern Medical Center, Dallas, Texas, USA.,Hamon Center for Regenerative Science and Medicine, The University of Texas Southwestern Medical Center, Dallas, Texas, USA
| |
Collapse
|
18
|
The cellular and molecular basis of somatosensory neuron development. Neuron 2021; 109:3736-3757. [PMID: 34592169 DOI: 10.1016/j.neuron.2021.09.004] [Citation(s) in RCA: 63] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Revised: 08/23/2021] [Accepted: 09/01/2021] [Indexed: 11/23/2022]
Abstract
Primary somatosensory neurons convey salient information about our external environment and internal state to the CNS, allowing us to detect, perceive, and react to a wide range of innocuous and noxious stimuli. Pseudo-unipolar in shape, and among the largest (longest) cells of most mammals, dorsal root ganglia (DRG) somatosensory neurons have peripheral axons that extend into skin, muscle, viscera, or bone and central axons that innervate the spinal cord and brainstem, where they synaptically engage the central somatosensory circuitry. Here, we review the diversity of mammalian DRG neuron subtypes and the intrinsic and extrinsic mechanisms that control their development. We describe classical and contemporary advances that frame our understanding of DRG neurogenesis, transcriptional specification of DRG neurons, and the establishment of morphological, physiological, and synaptic diversification across somatosensory neuron subtypes.
Collapse
|
19
|
García-Martínez FJ, Azorín D, Duat-Rodríguez A, Hernández-Martín Á. Angeborene kutane Neurofibrome bei Neurofibromatose Typ 1: Klinisch‐pathologische Merkmale in der frühen Kindheit. J Dtsch Dermatol Ges 2021; 19:73-81. [PMID: 33491906 DOI: 10.1111/ddg.14322_g] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Accepted: 05/26/2020] [Indexed: 01/21/2023]
Affiliation(s)
| | - Daniel Azorín
- Abteilung Pathologie, Hospital Infantil Universitario Niño Jesús, Madrid, Spanien
| | - Anna Duat-Rodríguez
- Abteilung Pädiatrische Neurologie, Hospital Infantil Universitario Niño Jesús, Madrid, Spanien
| | | |
Collapse
|
20
|
García-Martínez FJ, Azorín D, Duat-Rodríguez A, Hernández-Martín Á. Congenital cutaneous neurofibromas in neurofibromatosis type 1: Clinicopathological features in early infancy. J Dtsch Dermatol Ges 2021; 19:73-80. [PMID: 33448128 DOI: 10.1111/ddg.14322] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Accepted: 05/26/2020] [Indexed: 12/12/2022]
Abstract
BACKGROUND AND OBJECTIVE Clinicopathological features of cutaneous neurofibromas presenting as large irregularly shaped congenital café-au-lait macules (CALM) in Neurofibromatosis type 1 (NF1) patients have not been well characterized. We aimed to analyze the histopathological findings of large "atypical" CALM in children with NF1. PATIENTS AND METHODS In this retrospective observational study we analyzed histopathological and immunostaining features of 21 biopsy specimens from 18 large hyperpigmented macules with irregular borders with or without hypertrichosis present during the first months of life in NF1 diagnosed children. RESULTS Of the 21 biopsies, ten showed a diffuse neurofibroma pattern and four exhibited characteristics of plexiform neurofibroma (PNF). In twelve specimens we observed groups of fusiform cells arranged linearly mimicking a small caliber nerve trunk with abnormal morphology. Repeated biopsies from two of these lesions performed at different ages showed transformation to a plexiform pattern. An increased interstitial cellularity was observed in 17 samples that was more evident around eccrine glands in 16 or accompanying hair follicles and vascular structures in twelve samples. All these cells had immunoreactivity for S100-protein, CD68 and were Melan-A positive in 15 samples. CONCLUSION Clinicopathological findings of congenital cutaneous neurofibromas provide early diagnostic clues of NF1 with high relevance for monitoring of these patients.
Collapse
Affiliation(s)
| | - Daniel Azorín
- Pathology Department, Hospital Infantil Universitario Niño Jesús, Madrid, Spain
| | - Anna Duat-Rodríguez
- Pediatric Neurology Department, Hospital Infantil Universitario Niño Jesús, Madrid, Spain
| | | |
Collapse
|
21
|
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
|
22
|
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
|
23
|
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
|
24
|
Milichko V, Dyachuk V. Novel Glial Cell Functions: Extensive Potency, Stem Cell-Like Properties, and Participation in Regeneration and Transdifferentiation. Front Cell Dev Biol 2020; 8:809. [PMID: 33015034 PMCID: PMC7461986 DOI: 10.3389/fcell.2020.00809] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2020] [Accepted: 07/31/2020] [Indexed: 12/26/2022] Open
Abstract
Glial cells are the most abundant cells in both the peripheral and central nervous systems. During the past decade, a subpopulation of immature peripheral glial cells, namely, embryonic Schwann cell-precursors, have been found to perform important functions related to development. These cells have properties resembling those of the neural crest and, depending on their location in the body, can transform into several different cell types in peripheral tissues, including autonomic neurons. This review describes the multipotent properties of Schwann cell-precursors and their importance, together with innervation, during early development. The heterogeneity of Schwann cells, as revealed using single-cell transcriptomics, raises a question on whether some glial cells in the adult peripheral nervous system retain their stem cell-like properties. We also discuss how a deeper insight into the biology of both embryonic and adult Schwann cells might lead to an effective treatment of the damage of both neural and non-neural tissues, including the damage caused by neurodegenerative diseases. Furthermore, understanding the potential involvement of Schwann cells in the regulation of tumor development may reveal novel targets for cancer treatment.
Collapse
Affiliation(s)
- Valentin Milichko
- Department of Nanophotonics and Metamaterials, ITMO University, St. Petersburg, Russia
| | - Vyacheslav Dyachuk
- Department of Nanophotonics and Metamaterials, ITMO University, St. Petersburg, Russia.,National Scientific Center of Marine Biology, Far Eastern Branch, Russian Academy of Sciences, Vladivostok, Russia
| |
Collapse
|
25
|
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
|
26
|
Cobo R, García-Mesa Y, García-Piqueras J, Feito J, Martín-Cruces J, García-Suárez O, A. Vega J. The Glial Cell of Human Cutaneous Sensory Corpuscles: Origin, Characterization, and Putative Roles. Somatosens Mot Res 2020. [DOI: 10.5772/intechopen.91815] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
27
|
Schwarzer S, Asokan N, Bludau O, Chae J, Kuscha V, Kaslin J, Hans S. Neurogenesis in the inner ear: the zebrafish statoacoustic ganglion provides new neurons from a Neurod/Nestin-positive progenitor pool well into adulthood. Development 2020; 147:dev.176750. [PMID: 32165493 DOI: 10.1242/dev.176750] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Accepted: 02/25/2020] [Indexed: 01/13/2023]
Abstract
The vertebrate inner ear employs sensory hair cells and neurons to mediate hearing and balance. In mammals, damaged hair cells and neurons are not regenerated. In contrast, hair cells in the inner ear of zebrafish are produced throughout life and regenerate after trauma. However, it is unknown whether new sensory neurons are also formed in the adult zebrafish statoacoustic ganglion (SAG), the sensory ganglion connecting the inner ear to the brain. Using transgenic lines and marker analysis, we identify distinct cell populations and anatomical landmarks in the juvenile and adult SAG. In particular, we analyze a Neurod/Nestin-positive progenitor pool that produces large amounts of new neurons at juvenile stages, which transitions to a quiescent state in the adult SAG. Moreover, BrdU pulse chase experiments reveal the existence of a proliferative but otherwise marker-negative cell population that replenishes the Neurod/Nestin-positive progenitor pool at adult stages. Taken together, our study represents the first comprehensive characterization of the adult zebrafish SAG showing that zebrafish, in sharp contrast to mammals, display continued neurogenesis in the SAG well beyond embryonic and larval stages.
Collapse
Affiliation(s)
- Simone Schwarzer
- Center for Regenerative Therapies Dresden (CRTD), Cluster of Excellence, Center for Molecular and Cellular Bioengineering, Technische Universität Dresden, Dresden, Germany
| | - Nandini Asokan
- Center for Regenerative Therapies Dresden (CRTD), Cluster of Excellence, Center for Molecular and Cellular Bioengineering, Technische Universität Dresden, Dresden, Germany
| | - Oliver Bludau
- Center for Regenerative Therapies Dresden (CRTD), Cluster of Excellence, Center for Molecular and Cellular Bioengineering, Technische Universität Dresden, Dresden, Germany
| | - Jeongeun Chae
- Center for Regenerative Therapies Dresden (CRTD), Cluster of Excellence, Center for Molecular and Cellular Bioengineering, Technische Universität Dresden, Dresden, Germany
| | - Veronika Kuscha
- Center for Regenerative Therapies Dresden (CRTD), Cluster of Excellence, Center for Molecular and Cellular Bioengineering, Technische Universität Dresden, Dresden, Germany
| | - Jan Kaslin
- Center for Regenerative Therapies Dresden (CRTD), Cluster of Excellence, Center for Molecular and Cellular Bioengineering, Technische Universität Dresden, Dresden, Germany
| | - Stefan Hans
- Center for Regenerative Therapies Dresden (CRTD), Cluster of Excellence, Center for Molecular and Cellular Bioengineering, Technische Universität Dresden, Dresden, Germany
| |
Collapse
|
28
|
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
|
29
|
Brosseau JP, Pichard DC, Legius EH, Wolkenstein P, Lavker RM, Blakeley JO, Riccardi VM, Verma SK, Brownell I, Le LQ. The biology of cutaneous neurofibromas: Consensus recommendations for setting research priorities. Neurology 2019; 91:S14-S20. [PMID: 29987131 DOI: 10.1212/wnl.0000000000005788] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2017] [Accepted: 04/09/2018] [Indexed: 12/23/2022] Open
Abstract
OBJECTIVE A group of experts in dermatology, genetics, neuroscience, and regenerative medicine collaborated to summarize current knowledge on the defined factors contributing to cutaneous neurofibroma (cNF) development and to provide consensus recommendations for future research priorities to gain an improved understanding of the biology of cNF. METHODS The group members reviewed published and unpublished data on cNF and related diseases via literature search, defined a set of key topic areas deemed critical in cNF pathogenesis, and developed recommendations in a series of consensus meetings. RESULTS Five specific topic areas were identified as being relevant to providing an enhanced understanding of the biology of cNF: (1) defining the human cells of origin; (2) understanding the role of the microenvironment, focusing on neurons, mast cells, and fibroblasts; (3) defining the genetic and molecular differences between the cNFs, focusing on size and number; (4) understanding if sex hormones are critical for cNF development or progression; and (5) identifying challenges in establishing in vitro and in vivo models representing human cNF. CONCLUSIONS The complexity of cNF biology stems from its heterogeneity at multiple levels including genetic, spatial involvement, temporal development, and cellular composition. We propose a unified working model for cNF that builds a framework to address the key questions about cNF that, when answered, will provide the necessary understanding of cNF biology to allow meaningful development of therapies.
Collapse
Affiliation(s)
- Jean-Philippe Brosseau
- From the Department of Dermatology (J.P.B., L.Q.L.), UT Southwestern Medical Center, Dallas, TX; Dermatology Branch (D.C.P., I.B.), Center for Cancer Research, National Cancer Institutes of Health, Bethesda, MD; Human Genetics Department (E.H.L.), University of Leuven, Belgium; Division Cancer Immunity Transplantation Infections (P.W.), Paris Est Créteil University, France; Department of Dermatology (R.M.L.), Northwestern University, Chicago, IL; Department of Neurology (J.O.B., S.K.V.), The Neurofibromatosis Therapeutic Acceleration Program, The Johns Hopkins University School of Medicine, Baltimore, MD; and The NF Institute (V.M.R.), La Crescenta, CA
| | - Dominique C Pichard
- From the Department of Dermatology (J.P.B., L.Q.L.), UT Southwestern Medical Center, Dallas, TX; Dermatology Branch (D.C.P., I.B.), Center for Cancer Research, National Cancer Institutes of Health, Bethesda, MD; Human Genetics Department (E.H.L.), University of Leuven, Belgium; Division Cancer Immunity Transplantation Infections (P.W.), Paris Est Créteil University, France; Department of Dermatology (R.M.L.), Northwestern University, Chicago, IL; Department of Neurology (J.O.B., S.K.V.), The Neurofibromatosis Therapeutic Acceleration Program, The Johns Hopkins University School of Medicine, Baltimore, MD; and The NF Institute (V.M.R.), La Crescenta, CA
| | - Eric H Legius
- From the Department of Dermatology (J.P.B., L.Q.L.), UT Southwestern Medical Center, Dallas, TX; Dermatology Branch (D.C.P., I.B.), Center for Cancer Research, National Cancer Institutes of Health, Bethesda, MD; Human Genetics Department (E.H.L.), University of Leuven, Belgium; Division Cancer Immunity Transplantation Infections (P.W.), Paris Est Créteil University, France; Department of Dermatology (R.M.L.), Northwestern University, Chicago, IL; Department of Neurology (J.O.B., S.K.V.), The Neurofibromatosis Therapeutic Acceleration Program, The Johns Hopkins University School of Medicine, Baltimore, MD; and The NF Institute (V.M.R.), La Crescenta, CA
| | - Pierre Wolkenstein
- From the Department of Dermatology (J.P.B., L.Q.L.), UT Southwestern Medical Center, Dallas, TX; Dermatology Branch (D.C.P., I.B.), Center for Cancer Research, National Cancer Institutes of Health, Bethesda, MD; Human Genetics Department (E.H.L.), University of Leuven, Belgium; Division Cancer Immunity Transplantation Infections (P.W.), Paris Est Créteil University, France; Department of Dermatology (R.M.L.), Northwestern University, Chicago, IL; Department of Neurology (J.O.B., S.K.V.), The Neurofibromatosis Therapeutic Acceleration Program, The Johns Hopkins University School of Medicine, Baltimore, MD; and The NF Institute (V.M.R.), La Crescenta, CA
| | - Robert M Lavker
- From the Department of Dermatology (J.P.B., L.Q.L.), UT Southwestern Medical Center, Dallas, TX; Dermatology Branch (D.C.P., I.B.), Center for Cancer Research, National Cancer Institutes of Health, Bethesda, MD; Human Genetics Department (E.H.L.), University of Leuven, Belgium; Division Cancer Immunity Transplantation Infections (P.W.), Paris Est Créteil University, France; Department of Dermatology (R.M.L.), Northwestern University, Chicago, IL; Department of Neurology (J.O.B., S.K.V.), The Neurofibromatosis Therapeutic Acceleration Program, The Johns Hopkins University School of Medicine, Baltimore, MD; and The NF Institute (V.M.R.), La Crescenta, CA
| | - Jaishri O Blakeley
- From the Department of Dermatology (J.P.B., L.Q.L.), UT Southwestern Medical Center, Dallas, TX; Dermatology Branch (D.C.P., I.B.), Center for Cancer Research, National Cancer Institutes of Health, Bethesda, MD; Human Genetics Department (E.H.L.), University of Leuven, Belgium; Division Cancer Immunity Transplantation Infections (P.W.), Paris Est Créteil University, France; Department of Dermatology (R.M.L.), Northwestern University, Chicago, IL; Department of Neurology (J.O.B., S.K.V.), The Neurofibromatosis Therapeutic Acceleration Program, The Johns Hopkins University School of Medicine, Baltimore, MD; and The NF Institute (V.M.R.), La Crescenta, CA
| | - Vincent M Riccardi
- From the Department of Dermatology (J.P.B., L.Q.L.), UT Southwestern Medical Center, Dallas, TX; Dermatology Branch (D.C.P., I.B.), Center for Cancer Research, National Cancer Institutes of Health, Bethesda, MD; Human Genetics Department (E.H.L.), University of Leuven, Belgium; Division Cancer Immunity Transplantation Infections (P.W.), Paris Est Créteil University, France; Department of Dermatology (R.M.L.), Northwestern University, Chicago, IL; Department of Neurology (J.O.B., S.K.V.), The Neurofibromatosis Therapeutic Acceleration Program, The Johns Hopkins University School of Medicine, Baltimore, MD; and The NF Institute (V.M.R.), La Crescenta, CA
| | - Sharad K Verma
- From the Department of Dermatology (J.P.B., L.Q.L.), UT Southwestern Medical Center, Dallas, TX; Dermatology Branch (D.C.P., I.B.), Center for Cancer Research, National Cancer Institutes of Health, Bethesda, MD; Human Genetics Department (E.H.L.), University of Leuven, Belgium; Division Cancer Immunity Transplantation Infections (P.W.), Paris Est Créteil University, France; Department of Dermatology (R.M.L.), Northwestern University, Chicago, IL; Department of Neurology (J.O.B., S.K.V.), The Neurofibromatosis Therapeutic Acceleration Program, The Johns Hopkins University School of Medicine, Baltimore, MD; and The NF Institute (V.M.R.), La Crescenta, CA
| | - Isaac Brownell
- From the Department of Dermatology (J.P.B., L.Q.L.), UT Southwestern Medical Center, Dallas, TX; Dermatology Branch (D.C.P., I.B.), Center for Cancer Research, National Cancer Institutes of Health, Bethesda, MD; Human Genetics Department (E.H.L.), University of Leuven, Belgium; Division Cancer Immunity Transplantation Infections (P.W.), Paris Est Créteil University, France; Department of Dermatology (R.M.L.), Northwestern University, Chicago, IL; Department of Neurology (J.O.B., S.K.V.), The Neurofibromatosis Therapeutic Acceleration Program, The Johns Hopkins University School of Medicine, Baltimore, MD; and The NF Institute (V.M.R.), La Crescenta, CA
| | - Lu Q Le
- From the Department of Dermatology (J.P.B., L.Q.L.), UT Southwestern Medical Center, Dallas, TX; Dermatology Branch (D.C.P., I.B.), Center for Cancer Research, National Cancer Institutes of Health, Bethesda, MD; Human Genetics Department (E.H.L.), University of Leuven, Belgium; Division Cancer Immunity Transplantation Infections (P.W.), Paris Est Créteil University, France; Department of Dermatology (R.M.L.), Northwestern University, Chicago, IL; Department of Neurology (J.O.B., S.K.V.), The Neurofibromatosis Therapeutic Acceleration Program, The Johns Hopkins University School of Medicine, Baltimore, MD; and The NF Institute (V.M.R.), La Crescenta, CA.
| |
Collapse
|
30
|
Dai R, Hua W, Chen W, Xiong L, Li L, Li Y. Isolation, Characterization, and Safety Evaluation of Human Skin-Derived Precursors from an Adherent Monolayer Culture System. Stem Cells Int 2019; 2019:9194560. [PMID: 31531027 PMCID: PMC6721512 DOI: 10.1155/2019/9194560] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Revised: 06/18/2019] [Accepted: 07/16/2019] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND Skin-derived precursors (SKPs) are promising candidates for regenerative medicine. Several studies have transcultured human SKPs (termed tSKPs) from fibroblasts (FBs) expanded in monolayer culture. Herein, we optimized the procedure by treating flasks with poly-2-hydroxyethyl methacrylate (poly-HEMA). METHODS tSKPs generated from our adherent monolayer culture system were investigated for protein expression and differentiation capacity. The aggregated cells and the proliferative cells within tSKP spheres were detected by mix-culturing FBs expressing two different fluorescent proteins and BrdU- or EdU-positive cells, respectively. To distinguish tSKPs from FBs, we compared their phenotypes and transcriptomes. The tumorigenicity of tSKPs and FBs was also assessed in our study. RESULTS tSKPs expressed Versican, Fibronectin, Vimentin, Sox2, and Nestin. Under appropriate stimuli, tSKPs could differentiate to mesenchymal or neural lineages. While these spheres were heterogeneous populations consisting of both proliferative and aggregated cells, the rate of proliferative cells correlated with a seeding density. tSKPs, isolated from FBs, were distinctive from FBs in cell cycle, marker expression, neural differentiation potential, and transcript profiles despite the two sharing partial similarity in certain properties. As for tumorigenesis, both tSKPs and FBs could be considered as nontumorigenic ex vivo and in vivo. CONCLUSION tSKPs were heterogeneous populations presenting similar characteristics as traditional SKPs, while being different from FBs. The potential mixture of FBs in spheres did not affect the biosafety of tSKPs, as both of which had normal karyotype and nontumorigenicity. Taken together, we suggested tSKPs had potential applications in regenerative medicine.
Collapse
Affiliation(s)
- Ru Dai
- Department of Dermatology, Ningbo First Hospital, Zhejiang University, No. 59, Liuting Street, Ningbo, Zhejiang 315010, China
- Department of Dermatology, West China Hospital, Sichuan University, No. 37, Guo Xue Xiang, Chengdu, Sichuan 610041, China
| | - Wei Hua
- Department of Dermatology, West China Hospital, Sichuan University, No. 37, Guo Xue Xiang, Chengdu, Sichuan 610041, China
| | - Wei Chen
- Department of Dermatology, West China Hospital, Sichuan University, No. 37, Guo Xue Xiang, Chengdu, Sichuan 610041, China
| | - Lidan Xiong
- Department of Dermatology, West China Hospital, Sichuan University, No. 37, Guo Xue Xiang, Chengdu, Sichuan 610041, China
| | - Li Li
- Department of Dermatology, West China Hospital, Sichuan University, No. 37, Guo Xue Xiang, Chengdu, Sichuan 610041, China
| | - Yiming Li
- Department of Dermatology, West China Hospital, Sichuan University, No. 37, Guo Xue Xiang, Chengdu, Sichuan 610041, China
| |
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
|
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]
|
33
|
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]
|
34
|
Dupin E, Calloni GW, Coelho-Aguiar JM, Le Douarin NM. The issue of the multipotency of the neural crest cells. Dev Biol 2018; 444 Suppl 1:S47-S59. [DOI: 10.1016/j.ydbio.2018.03.024] [Citation(s) in RCA: 67] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2017] [Revised: 03/29/2018] [Accepted: 03/30/2018] [Indexed: 12/25/2022]
|
35
|
Radomska KJ, Coulpier F, Gresset A, Schmitt A, Debbiche A, Lemoine S, Wolkenstein P, Vallat JM, Charnay P, Topilko P. Cellular Origin, Tumor Progression, and Pathogenic Mechanisms of Cutaneous Neurofibromas Revealed by Mice with Nf1 Knockout in Boundary Cap Cells. Cancer Discov 2018; 9:130-147. [PMID: 30348676 DOI: 10.1158/2159-8290.cd-18-0156] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2018] [Revised: 05/09/2018] [Accepted: 09/19/2018] [Indexed: 11/16/2022]
Abstract
Patients carrying an inactive NF1 allele develop tumors of Schwann cell origin called neurofibromas (NF). Genetically engineered mouse models have significantly enriched our understanding of plexiform forms of NFs (pNF). However, this has not been the case for cutaneous neurofibromas (cNF), observed in all NF1 patients, as no previous model recapitulates their development. Here, we show that conditional Nf1 inactivation in Prss56-positive boundary cap cells leads to bona fide pNFs and cNFs. This work identifies subepidermal glia as a likely candidate for the cellular origin of cNFs and provides insights on disease mechanisms, revealing a long, multistep pathologic process in which inflammation-related signals play a pivotal role. This new mouse model is an important asset for future clinical and therapeutic investigations of NF1-associated neurofibromas. SIGNIFICANCE: Patients affected by NF1 develop numerous cNFs. We present a mouse model that faithfully recapitulates cNFs, identify a candidate cell type at their origin, analyze the steps involved in their formation, and show that their development is dramatically accelerated by skin injury. These findings have important clinical/therapeutic implications.This article is highlighted in the In This Issue feature, p. 1.
Collapse
Affiliation(s)
- Katarzyna J Radomska
- Ecole Normale Supérieure, PSL Research University, CNRS, Inserm, Institut de Biologie de l'Ecole Normale Supérieure (IBENS), Paris, France
| | - Fanny Coulpier
- Ecole Normale Supérieure, PSL Research University, CNRS, Inserm, Institut de Biologie de l'Ecole Normale Supérieure (IBENS), Paris, France
| | - Aurelie Gresset
- Ecole Normale Supérieure, PSL Research University, CNRS, Inserm, Institut de Biologie de l'Ecole Normale Supérieure (IBENS), Paris, France
| | - Alain Schmitt
- Institut Cochin, Inserm, CNRS, Université Paris Descartes, Paris, France
| | - Amal Debbiche
- Ecole Normale Supérieure, PSL Research University, CNRS, Inserm, Institut de Biologie de l'Ecole Normale Supérieure (IBENS), Paris, France
| | - Sophie Lemoine
- Genomic facility, Ecole Normale Supérieure, PSL Research University, CNRS, Inserm, Institut de Biologie de l'Ecole Normale Supérieure (IBENS), Paris, France
| | - Pierre Wolkenstein
- Département de Dermatologie, Centre de Référence des Neurofibromatoses, Hôpital Henri-Mondor, AP-HP, Créteil, France
| | - Jean-Michel Vallat
- National Reference Centre "Rare Peripheral Neuropathies," Department of Neurology, Centre Hospitalier Universitaire de Limoges, Limoges, France
| | - Patrick Charnay
- Ecole Normale Supérieure, PSL Research University, CNRS, Inserm, Institut de Biologie de l'Ecole Normale Supérieure (IBENS), Paris, France.
| | - Piotr Topilko
- Ecole Normale Supérieure, PSL Research University, CNRS, Inserm, Institut de Biologie de l'Ecole Normale Supérieure (IBENS), Paris, France.
| |
Collapse
|
36
|
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
|
37
|
Wittmann G, Lechan RM. Prss56 expression in the rodent hypothalamus: Inverse correlation with pro-opiomelanocortin suggests oscillatory gene expression in adult rat tanycytes. J Comp Neurol 2018; 526:2444-2461. [PMID: 30242838 DOI: 10.1002/cne.24504] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2018] [Revised: 06/27/2018] [Accepted: 07/12/2018] [Indexed: 11/11/2022]
Abstract
We recently reported that the number of hypothalamic tanycytes expressing pro-opiomelanocortin (Pomc) is highly variable among brains of adult rats. While its cause and significance remain unknown, identifying other variably expressed genes in tanycytes may help understand this curious phenomenon. In this in situ hybridization study, we report that the Prss56 gene, which encodes a trypsin-like serine protease and is expressed in neural stem/progenitor cells, shows a similarly variable mRNA expression in tanycytes of adult rats and correlates inversely with tanycyte Pomc mRNA. Prss56 was expressed in α1, β1, subsets of α2, and some median eminence γ tanycytes, but virtually absent from β2 tanycytes. Prss56 was also expressed in vimentin positive tanycyte-like cells in the parenchyma of the ventromedial and arcuate nuclei, and in thyrotropin beta subunit-expressing cells of the pars tuberalis of the pituitary. In contrast to adults, Prss56 expression was uniformly high in tanycytes in adolescent rats. In mice, Prss56-expressing tanycytes and parenchymal cells were also observed but fewer in number and without significant variations. The results identify Prss56 as a second gene that is expressed variably in tanycytes of adult rats. We propose that the variable, inversely correlating expression of Prss56 and Pomc reflect periodically oscillating gene expression in tanycytes rather than stable expression levels that vary between individual rats. A possible functional link between Prss56 and POMC, and Prss56 as a potential marker for migrating tanycytes are discussed.
Collapse
Affiliation(s)
- Gábor Wittmann
- Department of Medicine, Division of Endocrinology, Diabetes and Metabolism, Tupper Research Institute, Tufts Medical Center, Boston, Massachusetts
| | - Ronald M Lechan
- Department of Medicine, Division of Endocrinology, Diabetes and Metabolism, Tupper Research Institute, Tufts Medical Center, Boston, Massachusetts.,Department of Neuroscience, Tufts University School of Medicine, Boston, Massachusetts
| |
Collapse
|
38
|
Nichols EL, Green LA, Smith CJ. Ensheathing cells utilize dynamic tiling of neuronal somas in development and injury as early as neuronal differentiation. Neural Dev 2018; 13:19. [PMID: 30121077 PMCID: PMC6098834 DOI: 10.1186/s13064-018-0115-8] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2018] [Accepted: 07/27/2018] [Indexed: 12/18/2022] Open
Abstract
Background Glial cell ensheathment of specific components of neuronal circuits is essential for nervous system function. Although ensheathment of axonal segments of differentiated neurons has been investigated, ensheathment of neuronal cell somas, especially during early development when neurons are extending processes and progenitor populations are expanding, is still largely unknown. Methods To address this, we used time-lapse imaging in zebrafish during the initial formation of the dorsal root ganglia (DRG). Results Our results show that DRG neurons are ensheathed throughout their entire lifespan by a progenitor population. These ensheathing cells dynamically remodel during development to ensure axons can extend away from the neuronal cell soma into the CNS and out to the skin. As a population, ensheathing cells tile each DRG neuron to ensure neurons are tightly encased. In development and in experimental cell ablation paradigms, the oval shape of DRG neurons dynamically changes during partial unensheathment. During longer extended unensheathment neuronal soma shifting is observed. We further show the intimate relationship of these ensheathing cells with the neurons leads to immediate and choreographed responses to distal axonal damage to the neuron. Conclusion We propose that the ensheathing cells dynamically contribute to the shape and position of neurons in the DRG by their remodeling activity during development and are primed to dynamically respond to injury of the neuron. Electronic supplementary material The online version of this article (10.1186/s13064-018-0115-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Evan L Nichols
- Department of Biological Sciences, University of Notre Dame, 015 Galvin Life Sciences Building, Notre Dame, IN, 46556, USA
| | - Lauren A Green
- Department of Biological Sciences, University of Notre Dame, 015 Galvin Life Sciences Building, Notre Dame, IN, 46556, USA.,Center for Stem Cells and Regenerative Medicine, University of Notre Dame, Notre Dame, IN, USA
| | - Cody J Smith
- Department of Biological Sciences, University of Notre Dame, 015 Galvin Life Sciences Building, Notre Dame, IN, 46556, USA. .,Center for Stem Cells and Regenerative Medicine, University of Notre Dame, Notre Dame, IN, USA.
| |
Collapse
|
39
|
The Human Skin-Derived Precursors for Regenerative Medicine: Current State, Challenges, and Perspectives. Stem Cells Int 2018; 2018:8637812. [PMID: 30123295 PMCID: PMC6079335 DOI: 10.1155/2018/8637812] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2018] [Revised: 05/29/2018] [Accepted: 06/13/2018] [Indexed: 02/05/2023] Open
Abstract
Skin-derived precursors (SKPs) are an adult stem cell source with self-renewal and multipotent differentiation. Although rodent SKPs have been discussed in detail in substantial studies, human SKPs (hSKPs) are rarely reported. Understanding the biological properties and possible mechanisms underlying hSKPs has important implications for regenerative medicine particularly clinical applications, as human-derived sources are more suitable for clinical transplantation. The finding that hSKPs derivatives, such as neural and mesodermal progeny, have both in vitro and in vivo function without any genetical modification makes hSKPs a trustable, secure, and accessible resource for cell-based therapy. Here, we provide an overview of hSKPs, describing their characteristics, originations and niches, and potential applications. A comparison between traditional and innovative culture methods used for hSKPs is also introduced. Furthermore, we discuss the challenges and the future perspectives towards the field of hSKPs. With this review, we hope to point out the current stage of hSKPs and highlight the problems that remain in this field.
Collapse
|
40
|
Nascimento AI, Mar FM, Sousa MM. The intriguing nature of dorsal root ganglion neurons: Linking structure with polarity and function. Prog Neurobiol 2018; 168:86-103. [PMID: 29729299 DOI: 10.1016/j.pneurobio.2018.05.002] [Citation(s) in RCA: 88] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2017] [Revised: 04/26/2018] [Accepted: 05/01/2018] [Indexed: 11/26/2022]
Abstract
Dorsal root ganglion (DRG) neurons are the first neurons of the sensory pathway. They are activated by a variety of sensory stimuli that are then transmitted to the central nervous system. An important feature of DRG neurons is their unique morphology where a single process -the stem axon- bifurcates into a peripheral and a central axonal branch, with different functions and cellular properties. Distinctive structural aspects of the two DRG neuron branches may have important implications for their function in health and disease. However, the link between DRG axonal branch structure, polarity and function has been largely neglected in the field, and relevant information is rather scattered across the literature. In particular, ultrastructural differences between the two axonal branches are likely to account for the higher transport and regenerative ability of the peripheral DRG neuron axon when compared to the central one. Nevertheless, the cell intrinsic factors contributing to this central-peripheral asymmetry are still unknown. Here we critically review the factors that may underlie the functional asymmetry between the peripheral and central DRG axonal branches. Also, we discuss the hypothesis that DRG neurons may assemble a structure resembling the axon initial segment that may be responsible, at least in part, for their polarity and electrophysiological features. Ultimately, we suggest that the clarification of the axonal ultrastructure of DRG neurons using state-of-the-art techniques will be crucial to understand the physiology of this peculiar cell type.
Collapse
Affiliation(s)
- Ana Isabel Nascimento
- Nerve Regeneration Group, Instituto de Biologia Molecular e Celular-IBMC and Instituto de Inovação e Investigação em Saúde, University of Porto, Rua Alfredo Allen, 208, 4200-135 Porto, Portugal; Instituto de Ciências Biomédicas Abel Salazar-ICBAS, Rua Jorge Viterbo Ferreira 228, 4050-313 Porto, Portugal
| | - Fernando Milhazes Mar
- Nerve Regeneration Group, Instituto de Biologia Molecular e Celular-IBMC and Instituto de Inovação e Investigação em Saúde, University of Porto, Rua Alfredo Allen, 208, 4200-135 Porto, Portugal
| | - Mónica Mendes Sousa
- Nerve Regeneration Group, Instituto de Biologia Molecular e Celular-IBMC and Instituto de Inovação e Investigação em Saúde, University of Porto, Rua Alfredo Allen, 208, 4200-135 Porto, Portugal.
| |
Collapse
|
41
|
Izeta A. Comment on 'Adult skin-derived precursor Schwann cell grafts form growths in the injured spinal cord of Fischer rats'. ACTA ACUST UNITED AC 2018. [PMID: 29532787 DOI: 10.1088/1748-605x/aab628] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Affiliation(s)
- Ander Izeta
- Tissue Engineering Group, Bioengineering Area, Instituto Biodonostia, San Sebastian, E-20014, Spain. Department of Biomedical Engineering, School of Engineering, Tecnun-University of Navarra, San Sebastian, E-20009, Spain
| |
Collapse
|
42
|
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
|
43
|
Furlan A, Dyachuk V, Kastriti ME, Calvo-Enrique L, Abdo H, Hadjab S, Chontorotzea T, Akkuratova N, Usoskin D, Kamenev D, Petersen J, Sunadome K, Memic F, Marklund U, Fried K, Topilko P, Lallemend F, Kharchenko PV, Ernfors P, Adameyko I. Multipotent peripheral glial cells generate neuroendocrine cells of the adrenal medulla. Science 2018; 357:357/6346/eaal3753. [PMID: 28684471 DOI: 10.1126/science.aal3753] [Citation(s) in RCA: 225] [Impact Index Per Article: 32.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2016] [Accepted: 06/05/2017] [Indexed: 12/23/2022]
Abstract
Adrenaline is a fundamental circulating hormone for bodily responses to internal and external stressors. Chromaffin cells of the adrenal medulla (AM) represent the main neuroendocrine adrenergic component and are believed to differentiate from neural crest cells. We demonstrate that large numbers of chromaffin cells arise from peripheral glial stem cells, termed Schwann cell precursors (SCPs). SCPs migrate along the visceral motor nerve to the vicinity of the forming adrenal gland, where they detach from the nerve and form postsynaptic neuroendocrine chromaffin cells. An intricate molecular logic drives two sequential phases of gene expression, one unique for a distinct transient cellular state and another for cell type specification. Subsequently, these programs down-regulate SCP-gene and up-regulate chromaffin cell-gene networks. The AM forms through limited cell expansion and requires the recruitment of numerous SCPs. Thus, peripheral nerves serve as a stem cell niche for neuroendocrine system development.
Collapse
Affiliation(s)
- Alessandro Furlan
- Unit of Molecular Neurobiology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, 17177 Stockholm, Sweden
| | - Vyacheslav Dyachuk
- Department of Neuroscience, Karolinska Institutet, 17177 Stockholm, Sweden.,National Scientific Center of Marine Biology, Far Eastern Branch, Russian Academy of Sciences, Vladivostok 690041, Russia.,Department of Nanophotonics and Metamaterials, ITMO University, St. Petersburg 197101, Russia
| | - Maria Eleni Kastriti
- Department of Physiology and Pharmacology, Karolinska Institutet, 17177 Stockholm, Sweden
| | - Laura Calvo-Enrique
- Unit of Molecular Neurobiology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, 17177 Stockholm, Sweden
| | - Hind Abdo
- Unit of Molecular Neurobiology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, 17177 Stockholm, Sweden
| | - Saida Hadjab
- Department of Neuroscience, Karolinska Institutet, 17177 Stockholm, Sweden
| | - Tatiana Chontorotzea
- Department of Physiology and Pharmacology, Karolinska Institutet, 17177 Stockholm, Sweden
| | - Natalia Akkuratova
- Skolkovo Institute of Science and Technology, Moscow 143005, Russia.,Institute of Translational Biomedicine, Saint Petersburg State University, St. Petersburg 199034, Russia
| | - Dmitry Usoskin
- Unit of Molecular Neurobiology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, 17177 Stockholm, Sweden
| | - Dmitry Kamenev
- Department of Neuroscience, Karolinska Institutet, 17177 Stockholm, Sweden
| | - Julian Petersen
- Department of Physiology and Pharmacology, Karolinska Institutet, 17177 Stockholm, Sweden.,Center for Brain Research, Medical University Vienna, 1090 Vienna, Austria
| | - Kazunori Sunadome
- Department of Physiology and Pharmacology, Karolinska Institutet, 17177 Stockholm, Sweden
| | - Fatima Memic
- Unit of Molecular Neurobiology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, 17177 Stockholm, Sweden
| | - Ulrika Marklund
- Unit of Molecular Neurobiology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, 17177 Stockholm, Sweden
| | - Kaj Fried
- Department of Neuroscience, Karolinska Institutet, 17177 Stockholm, Sweden
| | - Piotr Topilko
- Institut de Biologie de l'Ecole Normale Supérieure, Ecole Normale Supérieure, INSERM U1024, CNRS UMR 8197, 46 Rue d'Ulm, 75005 Paris, France
| | - Francois Lallemend
- Department of Neuroscience, Karolinska Institutet, 17177 Stockholm, Sweden
| | - Peter V Kharchenko
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA 02115, USA
| | - Patrik Ernfors
- Unit of Molecular Neurobiology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, 17177 Stockholm, Sweden.
| | - Igor Adameyko
- Department of Physiology and Pharmacology, Karolinska Institutet, 17177 Stockholm, Sweden. .,Center for Brain Research, Medical University Vienna, 1090 Vienna, Austria
| |
Collapse
|
44
|
Kastriti ME, Adameyko I. Specification, plasticity and evolutionary origin of peripheral glial cells. Curr Opin Neurobiol 2017; 47:196-202. [DOI: 10.1016/j.conb.2017.11.004] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2017] [Revised: 08/30/2017] [Accepted: 11/03/2017] [Indexed: 12/31/2022]
|
45
|
Boundary cap cells in development and disease. Curr Opin Neurobiol 2017; 47:209-215. [DOI: 10.1016/j.conb.2017.11.003] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2017] [Revised: 10/10/2017] [Accepted: 11/03/2017] [Indexed: 01/18/2023]
|
46
|
Aquino JB, Sierra R. Schwann cell precursors in health and disease. Glia 2017; 66:465-476. [PMID: 29124786 DOI: 10.1002/glia.23262] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2017] [Revised: 10/07/2017] [Accepted: 10/26/2017] [Indexed: 12/25/2022]
Abstract
Schwann cell precursors (SCPs) are frequently regarded as neural crest-derived cells (NCDCs) found in contact with axons during nerve formation. Nevertheless, cells with SCPs properties can be found up to the adulthood. They are well characterized with regard to both gene expression profile and cellular behavior -for instance, proliferation, migratory capabilities and survival requirements-. They differ in origin regarding their anatomic location: even though most of them are derived from migratory NCCs, there is also contribution of the boundary cap neural crest cells (bNCCs) to the skin and other tissues. Many functions are known for SCPs in normal development, including nerve fasciculation and target innervation, arterial branching patterning and differentiation, and other morphogenetic processes. In addition, SCPs are now known to be a source of many neural (glia, endoneural fibroblasts, melanocytes, visceral neurons, and chromaffin cells) and non-neural-like (mesenchymal stromal cells, able e.g., to generate dentine-producing odontoblasts) cell types. Until now no reports of endoderm-like derivatives were reported so far. Interestingly, in the Schwann cell lineage only early SCPs are likely able to differentiate into melanocytes and bone marrow mesenchymal stromal cells. We have also herein discussed the literature regarding their role in repair as well as in disease mechanisms, such as in diverse cancers. Moreover, many caveats in our knowledge of SCPs biology are highlighted all through this article. Future research should expand more into the relevance of SCPs in pathologies and in other regenerative mechanisms which might bring new unexpected clinically-relevant knowledge.
Collapse
Affiliation(s)
- Jorge B Aquino
- Developmental Biology & Regenerative Medicine Laboratory, Instituto de Investigaciones en Medicina Traslacional (IIMT), CONICET-Universidad Austral, Derqui-Pilar, Buenos Aires, Argentina
| | - Romina Sierra
- Developmental Biology & Regenerative Medicine Laboratory, Instituto de Investigaciones en Medicina Traslacional (IIMT), CONICET-Universidad Austral, Derqui-Pilar, Buenos Aires, Argentina
| |
Collapse
|
47
|
Iribar H, Pérez-López V, Etxaniz U, Gutiérrez-Rivera A, Izeta A. Schwann Cells in the Ventral Dermis Do Not Derive from Myf5-Expressing Precursors. Stem Cell Reports 2017; 9:1477-1487. [PMID: 29033303 PMCID: PMC5830985 DOI: 10.1016/j.stemcr.2017.09.010] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2016] [Revised: 09/12/2017] [Accepted: 09/13/2017] [Indexed: 12/19/2022] Open
Abstract
The embryonic origin of lineage precursors of the trunk dermis is somewhat controversial. Precursor cells traced by Myf5 and Twist2 (Dermo1) promoter activation (i.e., cells of presumed dermomyotomal lineage) have been reported to generate Schwann cells. On the other hand, abundant data demonstrate that dermal Schwann cells derive from the neural crest. This is relevant because dermal precursors give rise to neural lineages, and multilineage differentiation potential qualifies them as adult stem cells. However, it is currently unclear whether neural lineages arise from dedifferentiated Schwann cells instead of mesodermally derived dermal precursor cells. To clarify these discrepancies, we traced SOX2+ adult dermal precursor cells by two independent Myf5 lineage tracing strains. We demonstrate that dermal Schwann cells do not belong to the Myf5+ cell lineage, indicating that previous tracing data reflected aberrant cre recombinase expression and that bona fide Myf5+ dermal precursors cannot transdifferentiate to neural lineages in physiological conditions. Adult Myf5-creSor mice aberrantly trace dermal Schwann cells (dSCs) Dedifferentiated, SOX2+ dSCs are the neural-competent precursors in the dermis These findings cast doubt on the multipotency of adult skin-derived precursors
Collapse
Affiliation(s)
- Haizea Iribar
- Tissue Engineering Laboratory, Bioengineering Area, Instituto Biodonostia, San Sebastian 20014, Spain
| | - Virginia Pérez-López
- Tissue Engineering Laboratory, Bioengineering Area, Instituto Biodonostia, San Sebastian 20014, Spain
| | - Usue Etxaniz
- Tissue Engineering Laboratory, Bioengineering Area, Instituto Biodonostia, San Sebastian 20014, Spain
| | - Araika Gutiérrez-Rivera
- Tissue Engineering Laboratory, Bioengineering Area, Instituto Biodonostia, San Sebastian 20014, Spain.
| | - Ander Izeta
- Tissue Engineering Laboratory, Bioengineering Area, Instituto Biodonostia, San Sebastian 20014, Spain; Department of Biomedical Engineering, School of Engineering, Tecnun-University of Navarra, San Sebastian 20009, Spain.
| |
Collapse
|
48
|
Livin' On The Edge: glia shape nervous system transition zones. Curr Opin Neurobiol 2017; 47:44-51. [PMID: 28957729 DOI: 10.1016/j.conb.2017.09.008] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Accepted: 09/11/2017] [Indexed: 11/21/2022]
Abstract
The vertebrate nervous system is divided into two functional halves; the central nervous system (CNS), which includes the brain and spinal cord, and the peripheral nervous system (PNS), which consists of nerves and ganglia. Incoming peripheral stimuli transmitted from the periphery to the CNS and subsequent motor responses created because of this information, require efficient communication between the two halves that make up this organ system. Neurons and glial cells of each half of the nervous system, which are the main actors in this communication, segregate across nervous system transition zones and never mix, allowing for efficient neurotransmission. Studies aimed at understanding the cellular and molecular mechanisms governing the development and maintenance of these transition zones have predominantly focused on mammalian models. However, zebrafish has emerged as a powerful model organism to study these structures and has allowed researchers to identify novel glial cells and mechanisms essential for nervous system assembly. This review will highlight recent advances into the important role that glial cells play in building and maintaining the nervous system and its boundaries.
Collapse
|
49
|
Zurkirchen L, Sommer L. Quo vadis: tracing the fate of neural crest cells. Curr Opin Neurobiol 2017; 47:16-23. [PMID: 28753439 DOI: 10.1016/j.conb.2017.07.001] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2017] [Revised: 06/30/2017] [Accepted: 07/03/2017] [Indexed: 12/16/2022]
Abstract
The neural crest is a transient structure in vertebrate embryos that produces migratory cells with an astonishing developmental potential. While neural crest fate maps have originally been established through interspecies transplantation assays, dye labeling, and retroviral infection, more recent methods rely on approaches involving transgenesis and genome editing. These technologies allowed the identification of minor neural crest-derived cell populations in tissues of non-neural crest origin. Furthermore, in vivo multipotency at the single cell level and stage-dependent fate acquisitions were demonstrated using genetic technologies. Finally, recent reports indicate that neural crest-derived cells become activated in response to injury to secrete factors supporting tissue repair. Thus, neural crest-derived cells apparently contribute to tissue formation and regeneration by cell autonomous and non-autonomous mechanisms.
Collapse
Affiliation(s)
- Luis Zurkirchen
- Stem Cell Biology, Institute of Anatomy, University of Zurich, Winterthurerstrasse 190, CH-8057 Zurich, Switzerland
| | - Lukas Sommer
- Stem Cell Biology, Institute of Anatomy, University of Zurich, Winterthurerstrasse 190, CH-8057 Zurich, Switzerland.
| |
Collapse
|
50
|
|