1
|
Birren SJ, Goodrich LV, Segal RA. Satellite Glial Cells: No Longer the Most Overlooked Glia. Cold Spring Harb Perspect Biol 2025; 17:a041367. [PMID: 38768970 PMCID: PMC11694750 DOI: 10.1101/cshperspect.a041367] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/22/2024]
Abstract
Many glial biologists consider glia the neglected cells of the nervous system. Among all the glia of the central and peripheral nervous system, satellite glia may be the most often overlooked. Satellite glial cells (SGCs) are located in ganglia of the cranial nerves and the peripheral nervous system. These small cells surround the cell bodies of neurons in the trigeminal ganglia (TG), spiral ganglia, nodose and petrosal ganglia, sympathetic ganglia, and dorsal root ganglia (DRG). Essential SGC features include their intimate connections with the associated neurons, their small size, and their derivation from neural crest cells. Yet SGCs also exhibit tissue-specific properties and can change rapidly, particularly in response to injury. To illustrate the range of SGC functions, we will focus on three types: those of the spiral, sympathetic, and DRG, and consider both their shared features and those that differ based on location.
Collapse
Affiliation(s)
- Susan J Birren
- Department of Biology, Brandeis University, Waltham, Massachusetts 02453, USA
| | - Lisa V Goodrich
- Department of Neurobiology, Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Rosalind A Segal
- Department of Neurobiology, Harvard Medical School, Boston, Massachusetts 02115, USA
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, Massachusetts 02215, USA
| |
Collapse
|
2
|
El-Hage O, Mikdache A, Boueid MJ, Degerny C, Tawk M. Schwann cells have a limited window of time in which to initiate myelination signaling during early migration in vivo. Cells Dev 2025:203993. [PMID: 39755275 DOI: 10.1016/j.cdev.2024.203993] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Revised: 11/29/2024] [Accepted: 12/28/2024] [Indexed: 01/06/2025]
Abstract
The temporal control of mitotic exit of individual Schwann cells (SCs) is essential for radial sorting and peripheral myelination. However, it remains unknown when, during their multiple rounds of division, SCs initiate myelin signaling in vivo. By manipulating SC division during development, we report that when SCs skip their division during migration, but not during radial sorting, they fail to myelinate peripheral axons. This coincides with a sharp decrease in Laminin expression within the posterior lateral line nerve. Interestingly, elevating cAMP levels or forcing Laminin 2 expression within individual SCs restore their ability to myelinate, despite missing mitosis during migration. Our results demonstrate a limited time window during which migrating SCs initiate Laminin expression to gradually activate the Laminin/Gpr126/cAMP signaling required for radial sorting and myelination at later stages in vivo.
Collapse
Affiliation(s)
- Océane El-Hage
- Université Paris-Saclay, Hôpital Kremlin Bicêtre, U1195, Inserm, 94276 Le Kremlin Bicêtre, France
| | - Aya Mikdache
- Université Paris-Saclay, Hôpital Kremlin Bicêtre, U1195, Inserm, 94276 Le Kremlin Bicêtre, France; UMR 3215 - U934, Institut Curie, 75005 Paris, France
| | - Marie-José Boueid
- Université Paris-Saclay, Hôpital Kremlin Bicêtre, U1195, Inserm, 94276 Le Kremlin Bicêtre, France
| | - Cindy Degerny
- Université Paris-Saclay, Hôpital Kremlin Bicêtre, U1195, Inserm, 94276 Le Kremlin Bicêtre, France.
| | - Marcel Tawk
- Université Paris-Saclay, Hôpital Kremlin Bicêtre, U1195, Inserm, 94276 Le Kremlin Bicêtre, France.
| |
Collapse
|
3
|
Chen Y, Shang T, Sun J, Ji Y, Gong L, Li A, Ding F, Shen M, Zhang Q. Characterization of sciatic nerve myelin sheath during development in C57BL/6 mice. Eur J Neurosci 2024; 60:4503-4517. [PMID: 38951719 DOI: 10.1111/ejn.16457] [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: 11/19/2023] [Revised: 06/05/2024] [Accepted: 06/16/2024] [Indexed: 07/03/2024]
Abstract
Myelin sheath plays important roles in information conduction and nerve injury repair in the peripheral nerve system (PNS). Enhancing comprehension of the structure and components of the myelin sheath in the PNS during development would contribute to a more comprehensive understanding of the developmental and regenerative processes. In this research, the structure of sciatic nerve myelin sheath in C57BL/6 mice from embryonic day 14 (E14) to postnatal 12 months (12M) was observed with transmission electron microscopy. Myelin structure appeared in the sciatic nerve as early as E14, and the number and thickness of myelin lamellar gradually increased with the development until 12M. Transcriptome analysis was performed to show the expressions of myelin-associated genes and transcriptional factors involved in myelin formation. The genes encoding myelin proteins (Mag, Pmp22, Mpz, Mbp, Cnp and Prx) showed the same expression pattern, peaking at postnatal day 7 (P7) and P28 after birth, whereas the negative regulators of myelination (c-Jun, Tgfb1, Tnc, Cyr61, Ngf, Egr1, Hgf and Bcl11a) showed an opposite expression pattern. In addition, the expression of myelin-associated proteins and transcriptional factors was measured by Western blot and immunofluorescence staining. The protein expressions of MAG, PMP22, MPZ, CNPase and PRX increased from E20 to P14. The key transcriptional factor c-Jun co-localized with the Schwann cells Marker S100β and decreased after birth, whereas Krox20/Egr2 increased during development. Our data characterized the structure and components of myelin sheath during the early developmental stages, providing insights for further understanding of PNS development.
Collapse
Affiliation(s)
- Yuhan Chen
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Medical School, Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Nantong University, Nantong, China
| | - Tongxin Shang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Medical School, Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Nantong University, Nantong, China
| | - Junjie Sun
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Medical School, Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Nantong University, Nantong, China
| | - Yuhua Ji
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Medical School, Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Nantong University, Nantong, China
| | - Leilei Gong
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Medical School, Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Nantong University, Nantong, China
- Research and Development Center for E-Learning, Ministry of Education, Beijing, China
| | - Aihong Li
- Department of Neurology, Affiliated Hospital of Nantong University, Medical School, Nantong University, Nantong, China
| | - Fei Ding
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Medical School, Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Nantong University, Nantong, China
| | - Mi Shen
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Medical School, Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Nantong University, Nantong, China
| | - Qi Zhang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Medical School, Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Nantong University, Nantong, China
| |
Collapse
|
4
|
Mrówczyńska E, Machalica K, Mazur AJ. Non-integrin laminin receptor (LamR) plays a role in axonal outgrowth from chicken DRG via modulating the Akt and Erk signaling. Front Cell Dev Biol 2024; 12:1433947. [PMID: 39144252 PMCID: PMC11322362 DOI: 10.3389/fcell.2024.1433947] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Accepted: 07/15/2024] [Indexed: 08/16/2024] Open
Abstract
37/67 kDa laminin receptor (LamR)/ribosomal protein SA exhibits dual function as both a ribosomal protein and cell surface receptor for laminin. LamR influences critical cellular processes such as invasion, adhesion, and migration when acting as a receptor. Despite the acknowledged importance of LamR/67LR in various cellular processes, its contribution to the peripheral nervous system development is obscure. Thus, this study investigated the biological activity of LamR in peripheral axonal outgrowth in the presence of laminin-1 or Ile-Lys-Val-Ala-Val (IKVAV) peptide, whose important role in dorsal root ganglia (DRG) axonal outgrowth we recently showed. Unexpectedly, we did not observe LamR on the surface of DRG cells or in a conditioned medium, suggesting its intracellular action in the negative regulation of DRG axonal outgrowth. Using C-terminus LamR-targeting IgG, we demonstrated the role of LamR in that process, which is independent of the presence of Schwann cell precursors (SCPs) and is mediated by extracellular signal-regulated kinase (Erk) and Protein kinase B (Akt1/2/3) signaling pathways. Additionally, we show that the action of LamR towards laminin-1-dependent axonal outgrowth is unmasked only when the activity of integrin β1 is perturbed. We believe that modulation of LamR activity provides the basis for its use for inhibiting axon growth as a potential therapeutic agent for regulating abnormal or excessive neurite growth during neurodevelopmental diseases or pathological nerve regeneration.
Collapse
Affiliation(s)
- Ewa Mrówczyńska
- Department of Cell Pathology, Faculty of Biotechnology, University of Wrocław, Wrocław, Poland
| | | | - Antonina Joanna Mazur
- Department of Cell Pathology, Faculty of Biotechnology, University of Wrocław, Wrocław, Poland
| |
Collapse
|
5
|
Takihira S, Yamada D, Osone T, Takao T, Sakaguchi M, Hakozaki M, Itano T, Nakata E, Fujiwara T, Kunisada T, Ozaki T, Takarada T. PRRX1-TOP2A interaction is a malignancy-promoting factor in human malignant peripheral nerve sheath tumours. Br J Cancer 2024; 130:1493-1504. [PMID: 38448751 PMCID: PMC11058259 DOI: 10.1038/s41416-024-02632-8] [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: 08/19/2023] [Revised: 02/15/2024] [Accepted: 02/19/2024] [Indexed: 03/08/2024] Open
Abstract
BACKGROUND Paired related-homeobox 1 (PRRX1) is a transcription factor in the regulation of developmental morphogenetic processes. There is growing evidence that PRRX1 is highly expressed in certain cancers and is critically involved in human survival prognosis. However, the molecular mechanism of PRRX1 in cancer malignancy remains to be elucidated. METHODS PRRX1 expression in human Malignant peripheral nerve sheath tumours (MPNSTs) samples was detected immunohistochemically to evaluate survival prognosis. MPNST models with PRRX1 gene knockdown or overexpression were constructed in vitro and the phenotype of MPNST cells was evaluated. Bioinformatics analysis combined with co-immunoprecipitation, mass spectrometry, RNA-seq and structural prediction were used to identify proteins interacting with PRRX1. RESULTS High expression of PRRX1 was associated with a poor prognosis for MPNST. PRRX1 knockdown suppressed the tumorigenic potential. PRRX1 overexpressed in MPNSTs directly interacts with topoisomerase 2 A (TOP2A) to cooperatively promote epithelial-mesenchymal transition and increase expression of tumour malignancy-related gene sets including mTORC1, KRAS and SRC signalling pathways. Etoposide, a TOP2A inhibitor used in the treatment of MPNST, may exhibit one of its anticancer effects by inhibiting the PRRX1-TOP2A interaction. CONCLUSION Targeting the PRRX1-TOP2A interaction in malignant tumours with high PRRX1 expression might provide a novel tumour-selective therapeutic strategy.
Collapse
Affiliation(s)
- Shota Takihira
- Department of Regenerative Science, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, 700-8558, Japan
- Department of Orthopedic Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, 700-8558, Japan
| | - Daisuke Yamada
- Department of Regenerative Science, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, 700-8558, Japan
| | - Tatsunori Osone
- Department of Regenerative Science, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, 700-8558, Japan
| | - Tomoka Takao
- Department of Regenerative Science, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, 700-8558, Japan
| | - Masakiyo Sakaguchi
- Department of Cell Biology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, 700-8558, Japan
| | - Michiyuki Hakozaki
- Department of Orthopedic Surgery, Fukushima Medical University School of Medicine, Fukushima, 960-1295, Japan
| | - Takuto Itano
- Department of Orthopedic Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, 700-8558, Japan
| | - Eiji Nakata
- Department of Orthopedic Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, 700-8558, Japan
| | - Tomohiro Fujiwara
- Department of Orthopedic Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, 700-8558, Japan
| | - Toshiyuki Kunisada
- Department of Orthopedic Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, 700-8558, Japan
| | - Toshifumi Ozaki
- Department of Orthopedic Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, 700-8558, Japan
| | - Takeshi Takarada
- Department of Regenerative Science, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, 700-8558, Japan.
| |
Collapse
|
6
|
Ayuso-García P, Sánchez-Rueda A, Velasco-Avilés S, Tamayo-Caro M, Ferrer-Pinós A, Huarte-Sebastian C, Alvarez V, Riobello C, Jiménez-Vega S, Buendia I, Cañas-Martin J, Fernández-Susavila H, Aparicio-Rey A, Esquinas-Román EM, Ponte CR, Guhl R, Laville N, Pérez-Andrés E, Lavín JL, González-Lopez M, Cámara NM, Aransay AM, Lozano JJ, Sutherland JD, Barrio R, Martinez-Chantar ML, Azkargorta M, Elortza F, Soriano-Navarro M, Matute C, Sánchez-Gómez MV, Bayón-Cordero L, Pérez-Samartín A, Bravo SB, Kurz T, Lama-Díaz T, Blanco MG, Haddad S, Record CJ, van Hasselt PM, Reilly MM, Varela-Rey M, Woodhoo A. Neddylation orchestrates the complex transcriptional and posttranscriptional program that drives Schwann cell myelination. SCIENCE ADVANCES 2024; 10:eadm7600. [PMID: 38608019 PMCID: PMC11014456 DOI: 10.1126/sciadv.adm7600] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/04/2023] [Accepted: 03/11/2024] [Indexed: 04/14/2024]
Abstract
Myelination is essential for neuronal function and health. In peripheral nerves, >100 causative mutations have been identified that cause Charcot-Marie-Tooth disease, a disorder that can affect myelin sheaths. Among these, a number of mutations are related to essential targets of the posttranslational modification neddylation, although how these lead to myelin defects is unclear. Here, we demonstrate that inhibiting neddylation leads to a notable absence of peripheral myelin and axonal loss both in developing and regenerating mouse nerves. Our data indicate that neddylation exerts a global influence on the complex transcriptional and posttranscriptional program by simultaneously regulating the expression and function of multiple essential myelination signals, including the master transcription factor EGR2 and the negative regulators c-Jun and Sox2, and inducing global secondary changes in downstream pathways, including the mTOR and YAP/TAZ signaling pathways. This places neddylation as a critical regulator of myelination and delineates the potential pathogenic mechanisms involved in CMT mutations related to neddylation.
Collapse
Affiliation(s)
- Paula Ayuso-García
- Gene Regulatory Control in Disease Laboratory, Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS), University of Santiago de Compostela, 15706 Santiago de Compostela, A Coruña, Spain
| | - Alejandro Sánchez-Rueda
- Gene Regulatory Control in Disease Laboratory, Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS), University of Santiago de Compostela, 15706 Santiago de Compostela, A Coruña, Spain
| | - Sergio Velasco-Avilés
- Gene Regulatory Control in Disease Laboratory, Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS), University of Santiago de Compostela, 15706 Santiago de Compostela, A Coruña, Spain
| | - Miguel Tamayo-Caro
- Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Bizkaia Technology Park, 48160 Derio, Spain
| | - Aroa Ferrer-Pinós
- Gene Regulatory Control in Disease Laboratory, Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS), University of Santiago de Compostela, 15706 Santiago de Compostela, A Coruña, Spain
| | - Cecilia Huarte-Sebastian
- Gene Regulatory Control in Disease Laboratory, Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS), University of Santiago de Compostela, 15706 Santiago de Compostela, A Coruña, Spain
| | - Vanesa Alvarez
- Gene Regulatory Control in Disease Laboratory, Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS), University of Santiago de Compostela, 15706 Santiago de Compostela, A Coruña, Spain
| | - Cristina Riobello
- Gene Regulatory Control in Disease Laboratory, Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS), University of Santiago de Compostela, 15706 Santiago de Compostela, A Coruña, Spain
| | - Selene Jiménez-Vega
- Gene Regulatory Control in Disease Laboratory, Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS), University of Santiago de Compostela, 15706 Santiago de Compostela, A Coruña, Spain
| | - Izaskun Buendia
- Gene Regulatory Control in Disease Laboratory, Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS), University of Santiago de Compostela, 15706 Santiago de Compostela, A Coruña, Spain
- Laboratory of Neurobiology, Achucarro Basque Center for Neuroscience, Science Park of UPV/EHU, Sede building, 48940 Leioa, Spain
| | - Jorge Cañas-Martin
- Gene Regulatory Control in Disease Laboratory, Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS), University of Santiago de Compostela, 15706 Santiago de Compostela, A Coruña, Spain
| | - Héctor Fernández-Susavila
- Gene Regulatory Control in Disease Laboratory, Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS), University of Santiago de Compostela, 15706 Santiago de Compostela, A Coruña, Spain
| | - Adrián Aparicio-Rey
- Gene Regulatory Control in Disease Laboratory, Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS), University of Santiago de Compostela, 15706 Santiago de Compostela, A Coruña, Spain
| | - Eva M. Esquinas-Román
- Gene Regulatory Control in Disease Laboratory, Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS), University of Santiago de Compostela, 15706 Santiago de Compostela, A Coruña, Spain
| | - Carlos Rodríguez Ponte
- Gene Regulatory Control in Disease Laboratory, Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS), University of Santiago de Compostela, 15706 Santiago de Compostela, A Coruña, Spain
| | - Romane Guhl
- Gene Regulatory Control in Disease Laboratory, Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS), University of Santiago de Compostela, 15706 Santiago de Compostela, A Coruña, Spain
- Université Paris Cité Magistère Européen de Génétique, 85 Boulevard Saint-Germain, 75006 Paris, France
| | - Nicolas Laville
- Gene Regulatory Control in Disease Laboratory, Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS), University of Santiago de Compostela, 15706 Santiago de Compostela, A Coruña, Spain
- Université Paris Cité Magistère Européen de Génétique, 85 Boulevard Saint-Germain, 75006 Paris, France
| | - Encarni Pérez-Andrés
- Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Bizkaia Technology Park, 48160 Derio, Spain
| | - José L. Lavín
- Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Bizkaia Technology Park, 48160 Derio, Spain
- NEIKER–Basque Institute for Agricultural Research and Development, Applied Mathematics Department, Bioinformatics Unit, Basque Research and Technology Alliance (BRTA), 48160 Derio, Bizkaia, Spain
| | - Monika González-Lopez
- Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Bizkaia Technology Park, 48160 Derio, Spain
| | - Nuria Macías Cámara
- Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Bizkaia Technology Park, 48160 Derio, Spain
| | - Ana M. Aransay
- Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Bizkaia Technology Park, 48160 Derio, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto de Salud Carlos III, Madrid, Spain
| | - Juan José Lozano
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto de Salud Carlos III, Madrid, Spain
| | - James D. Sutherland
- Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Bizkaia Technology Park, 48160 Derio, Spain
| | - Rosa Barrio
- Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Bizkaia Technology Park, 48160 Derio, Spain
| | - María Luz Martinez-Chantar
- Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Bizkaia Technology Park, 48160 Derio, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto de Salud Carlos III, Madrid, Spain
| | - Mikel Azkargorta
- Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Bizkaia Technology Park, 48160 Derio, Spain
| | - Félix Elortza
- Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Bizkaia Technology Park, 48160 Derio, Spain
| | - Mario Soriano-Navarro
- Electron Microscopy Core Facility, Centro de Investigación Príncipe Felipe (CIPF), 46012 Valencia, Spain
| | - Carlos Matute
- Laboratory of Neurobiology, Achucarro Basque Center for Neuroscience, Science Park of UPV/EHU, Sede building, 48940 Leioa, Spain
- Department of Neurosciences, University of the Basque Country (UPV/EHU), 48940 Leioa, Spain
- Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), 28031 Madrid, Spain
| | - María Victoria Sánchez-Gómez
- Laboratory of Neurobiology, Achucarro Basque Center for Neuroscience, Science Park of UPV/EHU, Sede building, 48940 Leioa, Spain
- Department of Neurosciences, University of the Basque Country (UPV/EHU), 48940 Leioa, Spain
- Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), 28031 Madrid, Spain
| | - Laura Bayón-Cordero
- Laboratory of Neurobiology, Achucarro Basque Center for Neuroscience, Science Park of UPV/EHU, Sede building, 48940 Leioa, Spain
- Department of Neurosciences, University of the Basque Country (UPV/EHU), 48940 Leioa, Spain
- Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), 28031 Madrid, Spain
| | - Alberto Pérez-Samartín
- Laboratory of Neurobiology, Achucarro Basque Center for Neuroscience, Science Park of UPV/EHU, Sede building, 48940 Leioa, Spain
- Department of Neurosciences, University of the Basque Country (UPV/EHU), 48940 Leioa, Spain
- Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), 28031 Madrid, Spain
| | - Susana B. Bravo
- Proteomic Unit, Health Research Institute of Santiago de Compostela (IDIS), 15705 Santiago de Compostela, A Coruña, Spain
| | - Thimo Kurz
- Evotec SE, Innovation Dr, Milton, Abingdon OX14 4RT, UK and School of Molecular Biosciences, University of Glasgow, Glasgow G12 8QQ, UK
| | - Tomas Lama-Díaz
- DNA Repair and Genome Integrity Laboratory, CIMUS, University of Santiago de Compostela-Instituto de Investigación Sanitaria, 15706 Santiago de Compostela, A Coruña, Spain
| | - Miguel G. Blanco
- DNA Repair and Genome Integrity Laboratory, CIMUS, University of Santiago de Compostela-Instituto de Investigación Sanitaria, 15706 Santiago de Compostela, A Coruña, Spain
- Department of Biochemistry and Molecular Biology, University of Santiago de Compostela, Plaza do Obradoiro s/n, Santiago de Compostela, Spain
| | - Saif Haddad
- Centre for Neuromuscular Diseases, UCL Queen Square Institute of Neurology, London, UK
| | - Christopher J. Record
- Centre for Neuromuscular Diseases, UCL Queen Square Institute of Neurology, London, UK
| | - Peter M. van Hasselt
- Department of Metabolic Diseases, Division Pediatrics, Wilhelmina Children’s Hospital University Medical Center Utrecht, Utrecht University, 3584 EA, Utrecht, Netherlands
| | - Mary M. Reilly
- Centre for Neuromuscular Diseases, UCL Queen Square Institute of Neurology, London, UK
| | - Marta Varela-Rey
- Gene Regulatory Control in Disease Laboratory, Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS), University of Santiago de Compostela, 15706 Santiago de Compostela, A Coruña, Spain
- Department of Biochemistry and Molecular Biology, University of Santiago de Compostela, Plaza do Obradoiro s/n, Santiago de Compostela, Spain
| | - Ashwin Woodhoo
- Gene Regulatory Control in Disease Laboratory, Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS), University of Santiago de Compostela, 15706 Santiago de Compostela, A Coruña, Spain
- Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Bizkaia Technology Park, 48160 Derio, Spain
- IKERBASQUE, Basque Foundation for Science, 48009 Bilbao, Bizkaia, Spain
- Department of Functional Biology, University of Santiago de Compostela, Plaza do Obradoiro s/n, Santiago de Compostela, Spain
- Oportunius Research Professor at CIMUS/USC, Galician Agency of Innovation (GAIN), Xunta de Galicia, Santiago de Compostela, A Coruña, Spain
| |
Collapse
|
7
|
Bai J, Yu B, Li C, Cheng H, Guan Y, Ren Z, Zhang T, Song X, Jia Z, Su T, Tao B, Gao H, Yang B, Liang L, Xiong X, Zhou X, Yin L, Peng J, Shang A, Wang Y. Mesenchymal Stem Cell-Derived Mitochondria Enhance Extracellular Matrix-Derived Grafts for the Repair of Nerve Defect. Adv Healthc Mater 2024; 13:e2302128. [PMID: 37922434 PMCID: PMC11468562 DOI: 10.1002/adhm.202302128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Revised: 10/27/2023] [Indexed: 11/05/2023]
Abstract
Peripheral nerve injuries (PNI) can lead to mitochondrial dysfunction and energy depletion within the affected microenvironment. The objective is to investigate the potential of transplanting mitochondria to reshape the neural regeneration microenvironment. High-purity functional mitochondria with an intact structure are extracted from human umbilical cord-derived mesenchymal stem cells (hUCMSCs) using the Dounce homogenization combined with ultracentrifugation. Results show that when hUCMSC-derived mitochondria (hUCMSC-Mitos) are cocultured with Schwann cells (SCs), they promote the proliferation, migration, and respiratory capacity of SCs. Acellular nerve allografts (ANAs) have shown promise in nerve regeneration, however, their therapeutic effect is not satisfactory enough. The incorporation of hUCMSC-Mitos within ANAs has the potential to remodel the regenerative microenvironment. This approach demonstrates satisfactory outcomes in terms of tissue regeneration and functional recovery. Particularly, the use of metabolomics and bioenergetic profiling is used for the first time to analyze the energy metabolism microenvironment after PNI. This remodeling occurs through the enhancement of the tricarboxylic acid cycle and the regulation of associated metabolites, resulting in increased energy synthesis. Overall, the hUCMSC-Mito-loaded ANAs exhibit high functionality to promote nerve regeneration, providing a novel regenerative strategy based on improving energy metabolism for neural repair.
Collapse
Affiliation(s)
- Jun Bai
- Department of NeurosurgeryGeneral Hospital of Chinese People Liberty ArmyNo. 28 Fuxing RoadBeijing100853P. R. China
- Institute of OrthopedicsThe Fourth Medical Center of Chinese PLA General Hospital; Beijing Key Lab of Regenerative Medicine in OrthopedicsKey Laboratory of Musculoskeletal Trauma and War Injuries PLANo. 51 Fucheng RoadBeijing100048P. R. China
- Co‐innovation Center of NeuroregenerationNantong University NantongJiangsu Province226007P. R. China
- Graduate School of Chinese PLA General HospitalNo. 28 Fuxing RoadBeijing100853P. R. China
| | - Bingbing Yu
- School of Materials Science and EngineeringThe Key Laboratory of Advanced Materials of Ministry of EducationState Key Laboratory of New Ceramics and Fine ProcessingCenter for Flexible Electronics TechnologyTsinghua UniversityBeijing100084P. R. China
| | - Chaochao Li
- Institute of OrthopedicsThe Fourth Medical Center of Chinese PLA General Hospital; Beijing Key Lab of Regenerative Medicine in OrthopedicsKey Laboratory of Musculoskeletal Trauma and War Injuries PLANo. 51 Fucheng RoadBeijing100048P. R. China
- Graduate School of Chinese PLA General HospitalNo. 28 Fuxing RoadBeijing100853P. R. China
| | - Haofeng Cheng
- Department of NeurosurgeryGeneral Hospital of Chinese People Liberty ArmyNo. 28 Fuxing RoadBeijing100853P. R. China
- Graduate School of Chinese PLA General HospitalNo. 28 Fuxing RoadBeijing100853P. R. China
- School of MedicineNankai UniversityTianjin300071P. R. China
| | - Yanjun Guan
- Institute of OrthopedicsThe Fourth Medical Center of Chinese PLA General Hospital; Beijing Key Lab of Regenerative Medicine in OrthopedicsKey Laboratory of Musculoskeletal Trauma and War Injuries PLANo. 51 Fucheng RoadBeijing100048P. R. China
- Graduate School of Chinese PLA General HospitalNo. 28 Fuxing RoadBeijing100853P. R. China
| | - Zhiqi Ren
- Department of NeurosurgeryGeneral Hospital of Chinese People Liberty ArmyNo. 28 Fuxing RoadBeijing100853P. R. China
- Graduate School of Chinese PLA General HospitalNo. 28 Fuxing RoadBeijing100853P. R. China
| | - Tieyuan Zhang
- Institute of OrthopedicsThe Fourth Medical Center of Chinese PLA General Hospital; Beijing Key Lab of Regenerative Medicine in OrthopedicsKey Laboratory of Musculoskeletal Trauma and War Injuries PLANo. 51 Fucheng RoadBeijing100048P. R. China
- Graduate School of Chinese PLA General HospitalNo. 28 Fuxing RoadBeijing100853P. R. China
| | - Xiangyu Song
- School of MedicineHebei North UniversityZhangjiakou075051P. R. China
| | - Zhibo Jia
- School of MedicineHebei North UniversityZhangjiakou075051P. R. China
| | - Tianqi Su
- Department of NeurosurgeryGeneral Hospital of Chinese People Liberty ArmyNo. 28 Fuxing RoadBeijing100853P. R. China
- Graduate School of Chinese PLA General HospitalNo. 28 Fuxing RoadBeijing100853P. R. China
| | - Benzhang Tao
- Department of NeurosurgeryGeneral Hospital of Chinese People Liberty ArmyNo. 28 Fuxing RoadBeijing100853P. R. China
| | - Haihao Gao
- Department of NeurosurgeryGeneral Hospital of Chinese People Liberty ArmyNo. 28 Fuxing RoadBeijing100853P. R. China
- Graduate School of Chinese PLA General HospitalNo. 28 Fuxing RoadBeijing100853P. R. China
| | - Boyao Yang
- Institute of OrthopedicsThe Fourth Medical Center of Chinese PLA General Hospital; Beijing Key Lab of Regenerative Medicine in OrthopedicsKey Laboratory of Musculoskeletal Trauma and War Injuries PLANo. 51 Fucheng RoadBeijing100048P. R. China
- Graduate School of Chinese PLA General HospitalNo. 28 Fuxing RoadBeijing100853P. R. China
| | - Lijing Liang
- Graduate School of Chinese PLA General HospitalNo. 28 Fuxing RoadBeijing100853P. R. China
| | - Xing Xiong
- Institute of OrthopedicsThe Fourth Medical Center of Chinese PLA General Hospital; Beijing Key Lab of Regenerative Medicine in OrthopedicsKey Laboratory of Musculoskeletal Trauma and War Injuries PLANo. 51 Fucheng RoadBeijing100048P. R. China
- Graduate School of Chinese PLA General HospitalNo. 28 Fuxing RoadBeijing100853P. R. China
| | - Xingyu Zhou
- Department of NeurosurgeryGeneral Hospital of Chinese People Liberty ArmyNo. 28 Fuxing RoadBeijing100853P. R. China
- Graduate School of Chinese PLA General HospitalNo. 28 Fuxing RoadBeijing100853P. R. China
| | - Lan Yin
- School of Materials Science and EngineeringThe Key Laboratory of Advanced Materials of Ministry of EducationState Key Laboratory of New Ceramics and Fine ProcessingCenter for Flexible Electronics TechnologyTsinghua UniversityBeijing100084P. R. China
| | - Jiang Peng
- Institute of OrthopedicsThe Fourth Medical Center of Chinese PLA General Hospital; Beijing Key Lab of Regenerative Medicine in OrthopedicsKey Laboratory of Musculoskeletal Trauma and War Injuries PLANo. 51 Fucheng RoadBeijing100048P. R. China
- Co‐innovation Center of NeuroregenerationNantong University NantongJiangsu Province226007P. R. China
| | - Aijia Shang
- Department of NeurosurgeryGeneral Hospital of Chinese People Liberty ArmyNo. 28 Fuxing RoadBeijing100853P. R. China
| | - Yu Wang
- Institute of OrthopedicsThe Fourth Medical Center of Chinese PLA General Hospital; Beijing Key Lab of Regenerative Medicine in OrthopedicsKey Laboratory of Musculoskeletal Trauma and War Injuries PLANo. 51 Fucheng RoadBeijing100048P. R. China
- Co‐innovation Center of NeuroregenerationNantong University NantongJiangsu Province226007P. R. China
| |
Collapse
|
8
|
Mohamed T, Melfi V, Colciago A, Magnaghi V. Hearing loss and vestibular schwannoma: new insights into Schwann cells implication. Cell Death Dis 2023; 14:629. [PMID: 37741837 PMCID: PMC10517973 DOI: 10.1038/s41419-023-06141-z] [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: 04/12/2023] [Revised: 09/05/2023] [Accepted: 09/08/2023] [Indexed: 09/25/2023]
Abstract
Hearing loss (HL) is the most common and heterogeneous disorder of the sensory system, with a large morbidity in the worldwide population. Among cells of the acoustic nerve (VIII cranial nerve), in the cochlea are present the hair cells, the spiral ganglion neurons, the glia-like supporting cells, and the Schwann cells (SCs), which alterations have been considered cause of HL. Notably, a benign SC-derived tumor of the acoustic nerve, named vestibular schwannoma (VS), has been indicated as cause of HL. Importantly, SCs are the main glial cells ensheathing axons and forming myelin in the peripheral nerves. Following an injury, the SCs reprogram, expressing some stemness features. Despite the mechanisms and factors controlling their biological processes (i.e., proliferation, migration, differentiation, and myelination) have been largely unveiled, their role in VS and HL was poorly investigated. In this review, we enlighten some of the mechanisms at the base of SCs transformation, VS development, and progression, likely leading to HL, and we pose great attention on the environmental factors that, in principle, could contribute to HL onset or progression. Combining the biomolecular bench-side approach to the clinical bedside practice may be helpful for the diagnosis, prediction, and therapeutic approach in otology.
Collapse
Affiliation(s)
- Tasnim Mohamed
- Department of Pharmacological and Biomolecular Sciences "Rodolfo Paoletti", Università degli Studi di Milano, Via G. Balzaretti 9, 20133, Milan, Italy
| | - Valentina Melfi
- Department of Pharmacological and Biomolecular Sciences "Rodolfo Paoletti", Università degli Studi di Milano, Via G. Balzaretti 9, 20133, Milan, Italy
- Fondazione IRCCS Istituto Nazionale dei Tumori, Via G. Venezian 1, 20133, Milan, Italy
| | - Alessandra Colciago
- Department of Pharmacological and Biomolecular Sciences "Rodolfo Paoletti", Università degli Studi di Milano, Via G. Balzaretti 9, 20133, Milan, Italy
| | - Valerio Magnaghi
- Department of Pharmacological and Biomolecular Sciences "Rodolfo Paoletti", Università degli Studi di Milano, Via G. Balzaretti 9, 20133, Milan, Italy.
| |
Collapse
|
9
|
Brown LN, Barth JL, Jafri S, Rumschlag JA, Jenkins TR, Atkinson C, Lang H. Complement factor B is essential for the proper function of the peripheral auditory system. Front Neurol 2023; 14:1214408. [PMID: 37560455 PMCID: PMC10408708 DOI: 10.3389/fneur.2023.1214408] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2023] [Accepted: 07/05/2023] [Indexed: 08/11/2023] Open
Abstract
Sensorineural hearing loss is associated with dysfunction of cochlear cells. Although immune cells play a critical role in maintaining the inner ear microenvironment, the precise immune-related molecular mechanisms underlying the pathophysiology of hearing loss remain unclear. The complement cascade contributes to the regulation of immune cell activity. Additionally, activation of the complement cascade can lead to the cellular opsonization of cells and pathogens, resulting in their engulfment and elimination by phagocytes. Complement factor B (fB) is an essential activator protein in the alternative complement pathway, and variations in the fB gene are associated with age-related macular degeneration. Here we show that mice of both sexes deficient in fB functional alleles (fB-/-) demonstrate progressive hearing impairment. Transcriptomic analysis of auditory nerves from adult mice detected 706 genes that were significantly differentially expressed between fB-/- and wild-type control animals, including genes related to the extracellular matrix and neural development processes. Additionally, a subset of differentially expressed genes was related to myelin function and neural crest development. Histological and immunohistochemical investigations revealed pathological alterations in auditory nerve myelin sheathes of fB-/- mice. Pathological alterations were also seen in the stria vascularis of the cochlear lateral wall in these mice. Our results implicate fB as an integral regulator of myelin maintenance and stria vascularis integrity, underscoring the importance of understanding the involvement of immune signaling pathways in sensorineural hearing loss.
Collapse
Affiliation(s)
- LaShardai N. Brown
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, SC, United States
| | - Jeremy L. Barth
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC, United States
| | - Shabih Jafri
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, SC, United States
| | - Jeffrey A. Rumschlag
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, SC, United States
| | - Tyreek R. Jenkins
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, SC, United States
| | - Carl Atkinson
- Division of Pulmonary Medicine, University of Florida, Gainesville, FL, United States
| | - Hainan Lang
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, SC, United States
| |
Collapse
|
10
|
Abstract
ABSTRACT Peripheral nerve injury is a common injury disease. Understanding of the mechanisms of periphery nerve repair and regeneration after injury is an essential prerequisite for treating related diseases. Although the biological mechanisms of peripheral nerve injury and regeneration have been studied comprehensively, the clinical treatment methods are still limited. The bottlenecks of the treatments are the shortage of donor nerves and the limited surgical precision. Apart from the knowledge regarding the fundamental characteristics and physical processes of peripheral nerve injury, numerous studies have found that Schwann cells, growth factors, and extracellular matrix are main factors affecting the repair and regeneration process of injured nerves. At present, the therapeutical methods of the disease include microsurgery, autologous nerve transplantation, allograft nerve transplantation and tissue engineering technology. Tissue engineering technology, which combines seed cells, neurotrophic factors, and scaffold materials together, is promising for treating the patients with long-gapped and large nerve damage. With the development of neuron science and technology, the treatment of peripheral nerve injury diseases will continue being improved.
Collapse
|
11
|
Abstract
Satellite glial cells (SGCs) that surround sensory neurons in the peripheral nervous system ganglia originate from neural crest cells. Although several studies have focused on SGCs, the origin and characteristics of SGCs are unknown, and their lineage remains unidentified. Traditionally, it has been considered that SGCs regulate the environment around neurons under pathological conditions, and perform functions of supporting, nourishing, and protecting neurons. However, recent studies demonstrated that SGCs may have the characteristics of stem cells. After nerve injury, SGCs up-regulate the expression of stem cell markers and can differentiate into functional sensory neurons. Moreover, SGCs express several markers of Schwann cell precursors and Schwann cells, such as CDH19, MPZ, PLP1, SOX10, ERBB3, and FABP7. Schwann cell precursors have also been proposed as a potential source of neurons in the peripheral nervous system. The similarity in function and markers suggests that SGCs may represent a subgroup of Schwann cell precursors. Herein, we discuss the roles and functions of SGCs, and the lineage relationship between SGCs and Schwann cell precursors. We also describe a new perspective on the roles and functions of SGCs. In the DRG located on the posterior root of spinal nerves, satellite glial cells wrap around each sensory neuron to form an anatomically and functionally distinct unit with the sensory neurons. Following nerve injury, satellite glial cells up-regulate the expression of progenitor markers, and can differentiate into neurons.
Collapse
|
12
|
Emerging Roles of Cholinergic Receptors in Schwann Cell Development and Plasticity. Biomedicines 2022; 11:biomedicines11010041. [PMID: 36672549 PMCID: PMC9855772 DOI: 10.3390/biomedicines11010041] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2022] [Revised: 12/12/2022] [Accepted: 12/20/2022] [Indexed: 12/29/2022] Open
Abstract
The cross talk between neurons and glial cells during development, adulthood, and disease, has been extensively documented. Among the molecules mediating these interactions, neurotransmitters play a relevant role both in myelinating and non-myelinating glial cells, thus resulting as additional candidates regulating the development and physiology of the glial cells. In this review, we summarise the contribution of the main neurotransmitter receptors in the regulation of the morphogenetic events of glial cells, with particular attention paid to the role of acetylcholine receptors in Schwann cell physiology. In particular, the M2 muscarinic receptor influences Schwann cell phenotype and the α7 nicotinic receptor is emerging as influential in the modulation of peripheral nerve regeneration and inflammation. This new evidence significantly improves our knowledge of Schwann cell development and function and may contribute to identifying interesting new targets to support the activity of these cells in pathological conditions.
Collapse
|
13
|
Cristobal CD, Lee HK. Development of myelinating glia: An overview. Glia 2022; 70:2237-2259. [PMID: 35785432 PMCID: PMC9561084 DOI: 10.1002/glia.24238] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Revised: 06/24/2022] [Accepted: 06/24/2022] [Indexed: 01/07/2023]
Abstract
Myelin is essential to nervous system function, playing roles in saltatory conduction and trophic support. Oligodendrocytes (OLs) and Schwann cells (SCs) form myelin in the central and peripheral nervous systems respectively and follow different developmental paths. OLs are neural stem-cell derived and follow an intrinsic developmental program resulting in a largely irreversible differentiation state. During embryonic development, OL precursor cells (OPCs) are produced in distinct waves originating from different locations in the central nervous system, with a subset developing into myelinating OLs. OPCs remain evenly distributed throughout life, providing a population of responsive, multifunctional cells with the capacity to remyelinate after injury. SCs derive from the neural crest, are highly dependent on extrinsic signals, and have plastic differentiation states. SC precursors (SCPs) are produced in early embryonic nerve structures and differentiate into multipotent immature SCs (iSCs), which initiate radial sorting and differentiate into myelinating and non-myelinating SCs. Differentiated SCs retain the capacity to radically change phenotypes in response to external signals, including becoming repair SCs, which drive peripheral regeneration. While several transcription factors and myelin components are common between OLs and SCs, their differentiation mechanisms are highly distinct, owing to their unique lineages and their respective environments. In addition, both OLs and SCs respond to neuronal activity and regulate nervous system output in reciprocal manners, possibly through different pathways. Here, we outline their basic developmental programs, mechanisms regulating their differentiation, and recent advances in the field.
Collapse
Affiliation(s)
- Carlo D. Cristobal
- Integrative Program in Molecular and Biomedical SciencesBaylor College of MedicineHoustonTexasUSA,Jan and Dan Duncan Neurological Research InstituteTexas Children's HospitalHoustonTexasUSA
| | - Hyun Kyoung Lee
- Integrative Program in Molecular and Biomedical SciencesBaylor College of MedicineHoustonTexasUSA,Jan and Dan Duncan Neurological Research InstituteTexas Children's HospitalHoustonTexasUSA,Department of PediatricsBaylor College of MedicineHoustonTexasUSA,Department of NeuroscienceBaylor College of MedicineHoustonTexasUSA
| |
Collapse
|
14
|
Mikdache A, Boueid MJ, Lesport E, Delespierre B, Loisel-Duwattez J, Degerny C, Tawk M. Timely Schwann cell division drives peripheral myelination in vivo via the laminin/cAMP pathway. Development 2022; 149:276236. [DOI: 10.1242/dev.200640] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Accepted: 07/29/2022] [Indexed: 11/20/2022]
Abstract
ABSTRACT
Schwann cells (SCs) migrate along peripheral axons and divide intensively to generate the right number of cells prior to axonal ensheathment; however, little is known regarding the temporal and molecular control of their division and its impact on myelination. We report that Sil, a spindle pole protein associated with autosomal recessive primary microcephaly, is required for temporal mitotic exit of SCs. In sil-deficient cassiopeia (csp−/−) mutants, SCs fail to radially sort and myelinate peripheral axons. Elevation of cAMP, but not Rac1 activity, in csp−/− restores myelin ensheathment. Most importantly, we show a significant decrease in laminin expression within csp−/− posterior lateral line nerve and that forcing Laminin 2 expression in csp−/− fully restores the ability of SCs to myelinate. Thus, we demonstrate an essential role for timely SC division in mediating laminin expression to orchestrate radial sorting and peripheral myelination in vivo.
Collapse
Affiliation(s)
- Aya Mikdache
- U1195, Inserm, University Paris-Saclay , 94276 Le Kremlin Bicêtre , France
| | - Marie-José Boueid
- U1195, Inserm, University Paris-Saclay , 94276 Le Kremlin Bicêtre , France
| | - Emilie Lesport
- U1195, Inserm, University Paris-Saclay , 94276 Le Kremlin Bicêtre , France
| | | | | | - Cindy Degerny
- U1195, Inserm, University Paris-Saclay , 94276 Le Kremlin Bicêtre , France
| | - Marcel Tawk
- U1195, Inserm, University Paris-Saclay , 94276 Le Kremlin Bicêtre , France
| |
Collapse
|
15
|
The Cell Autonomous and Non-Cell Autonomous Aspects of Neuronal Vulnerability and Resilience in Amyotrophic Lateral Sclerosis. BIOLOGY 2022; 11:biology11081191. [PMID: 36009818 PMCID: PMC9405388 DOI: 10.3390/biology11081191] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Revised: 07/14/2022] [Accepted: 07/30/2022] [Indexed: 11/17/2022]
Abstract
Simple Summary Amyotrophic lateral sclerosis (ALS) is a fatal disease characterized by a progressive paralysis due to the loss of particular neurons in our nervous system called motor neurons, that exert voluntary control of all our skeletal muscles. It is not entirely understood why motor neurons are particularly vulnerable in ALS, neither is it completely clear why certain groups of motor neurons, including those that regulate eye movement, are rather resilient to this disease. However, both vulnerability and resilience to ALS likely reflect cell intrinsic properties of different motor neuron subpopulations as well as non-cell autonomous events regulated by surrounding cell types. In this review we dissect the particular properties of different motor neuron types and their responses to disease that may underlie their respective vulnerabilities and resilience. Disease progression in ALS involves multiple cell types that are closely connected to motor neurons and we here also discuss their contributions to the differential vulnerability of motor neurons. Abstract Amyotrophic lateral sclerosis (ALS) is defined by the loss of upper motor neurons (MNs) that project from the cerebral cortex to the brain stem and spinal cord and of lower MNs in the brain stem and spinal cord which innervate skeletal muscles, leading to spasticity, muscle atrophy, and paralysis. ALS involves several disease stages, and multiple cell types show dysfunction and play important roles during distinct phases of disease initiation and progression, subsequently leading to selective MN loss. Why MNs are particularly vulnerable in this lethal disease is still not entirely clear. Neither is it fully understood why certain MNs are more resilient to degeneration in ALS than others. Brain stem MNs of cranial nerves III, IV, and VI, which innervate our eye muscles, are highly resistant and persist until the end-stage of the disease, enabling paralyzed patients to communicate through ocular tracking devices. MNs of the Onuf’s nucleus in the sacral spinal cord, that innervate sphincter muscles and control urogenital functions, are also spared throughout the disease. There is also a differential vulnerability among MNs that are intermingled throughout the spinal cord, that directly relate to their physiological properties. Here, fast-twitch fatigable (FF) MNs, which innervate type IIb muscle fibers, are affected early, before onset of clinical symptoms, while slow-twitch (S) MNs, that innervate type I muscle fibers, remain longer throughout the disease progression. The resilience of particular MN subpopulations has been attributed to intrinsic determinants and multiple studies have demonstrated their unique gene regulation and protein content in health and in response to disease. Identified factors within resilient MNs have been utilized to protect more vulnerable cells. Selective vulnerability may also, in part, be driven by non-cell autonomous processes and the unique surroundings and constantly changing environment close to particular MN groups. In this article, we review in detail the cell intrinsic properties of resilient and vulnerable MN groups, as well as multiple additional cell types involved in disease initiation and progression and explain how these may contribute to the selective MN resilience and vulnerability in ALS.
Collapse
|
16
|
Abstract
Schwann cells in the peripheral nervous system (PNS) are essential for the support and myelination of axons, ensuring fast and accurate communication between the central nervous system and the periphery. Schwann cells and related glia accompany innervating axons in virtually all tissues in the body, where they exhibit remarkable plasticity and the ability to modulate pathology in extraordinary, and sometimes surprising, ways. Here, we provide a brief overview of the various glial cell types in the PNS and describe the cornerstone cellular and molecular processes that enable Schwann cells to perform their canonical functions. We then dive into discussing exciting noncanonical functions of Schwann cells and related PNS glia, which include their role in organizing the PNS, in regulating synaptic activity and pain, in modulating immunity, in providing a pool of stem cells for different organs, and, finally, in influencing cancer.
Collapse
Affiliation(s)
- Carla Taveggia
- Axo-Glial Interaction Unit, Division of Neuroscience, IRCCS Ospedale San Raffaele, Milan, Italy;
| | - M. Laura Feltri
- Institute for Myelin and Glia Exploration, Departments of Biochemistry and Neurology, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, New York, USA
| |
Collapse
|
17
|
Fu H, Hu D, Chen J, Wang Q, Zhang Y, Qi C, Yu T. Repair of the Injured Spinal Cord by Schwann Cell Transplantation. Front Neurosci 2022; 16:800513. [PMID: 35250447 PMCID: PMC8891437 DOI: 10.3389/fnins.2022.800513] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2021] [Accepted: 01/27/2022] [Indexed: 01/12/2023] Open
Abstract
Spinal cord injury (SCI) can result in sensorimotor impairments or disability. Studies of the cellular response to SCI have increased our understanding of nerve regenerative failure following spinal cord trauma. Biological, engineering and rehabilitation strategies for repairing the injured spinal cord have shown impressive results in SCI models of both rodents and non-human primates. Cell transplantation, in particular, is becoming a highly promising approach due to the cells’ capacity to provide multiple benefits at the molecular, cellular, and circuit levels. While various cell types have been investigated, we focus on the use of Schwann cells (SCs) to promote SCI repair in this review. Transplantation of SCs promotes functional recovery in animal models and is safe for use in humans with subacute SCI. The rationales for the therapeutic use of SCs for SCI include enhancement of axon regeneration, remyelination of newborn or sparing axons, regulation of the inflammatory response, and maintenance of the survival of damaged tissue. However, little is known about the molecular mechanisms by which transplanted SCs exert a reparative effect on SCI. Moreover, SC-based therapeutic strategies face considerable challenges in preclinical studies. These issues must be clarified to make SC transplantation a feasible clinical option. In this review, we summarize the recent advances in SC transplantation for SCI, and highlight proposed mechanisms and challenges of SC-mediated therapy. The sparse information available on SC clinical application in patients with SCI is also discussed.
Collapse
Affiliation(s)
- Haitao Fu
- Department of Sports Medicine, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China
| | - Die Hu
- State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Qingdao Eye Hospital, Shandong Eye Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Qingdao, China
| | - Jinli Chen
- Department of Sports Medicine, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China
| | - Qizun Wang
- Department of Orthopedics, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Yingze Zhang
- Key Laboratory of Biomechanics of Hebei Province, Department of Trauma Emergency Center, The Third Hospital of Hebei Medical University, Orthopaedics Research Institution of Hebei Province, Shijiazhuang, China
| | - Chao Qi
- Department of Sports Medicine, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China
- *Correspondence: Chao Qi,
| | - Tengbo Yu
- Department of Sports Medicine, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China
- Tengbo Yu,
| |
Collapse
|
18
|
Modeling iPSC-derived human neurofibroma-like tumors in mice uncovers the heterogeneity of Schwann cells within plexiform neurofibromas. Cell Rep 2022; 38:110385. [PMID: 35172160 DOI: 10.1016/j.celrep.2022.110385] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Revised: 11/04/2021] [Accepted: 01/24/2022] [Indexed: 12/20/2022] Open
Abstract
Plexiform neurofibromas (pNFs) are developmental tumors that appear in neurofibromatosis type 1 individuals, constituting a major source of morbidity and potentially transforming into a highly metastatic sarcoma (MPNST). pNFs arise after NF1 inactivation in a cell of the neural crest (NC)-Schwann cell (SC) lineage. Here, we develop an iPSC-based NC-SC in vitro differentiation system and construct a lineage expression roadmap for the analysis of different 2D and 3D NF models. The best model consists of generating heterotypic spheroids (neurofibromaspheres) composed of iPSC-derived differentiating NF1(-/-) SCs and NF1(+/-) pNF-derived fibroblasts (Fbs). Neurofibromaspheres form by maintaining highly proliferative NF1(-/-) cells committed to the NC-SC axis due to SC-SC and SC-Fb interactions, resulting in SC linage cells at different maturation points. Upon engraftment on the mouse sciatic nerve, neurofibromaspheres consistently generate human NF-like tumors. Analysis of expression roadmap genes in human pNF single-cell RNA-seq data uncovers the presence of SC subpopulations at distinct differentiation states.
Collapse
|
19
|
Taïb S, Lamandé N, Martin S, Coulpier F, Topilko P, Brunet I. Myelinating Schwann cells and Netrin-1 control intra-nervous vascularization of the developing mouse sciatic nerve. eLife 2022; 11:64773. [PMID: 35019839 PMCID: PMC8782568 DOI: 10.7554/elife.64773] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Accepted: 01/11/2022] [Indexed: 11/13/2022] Open
Abstract
Peripheral nerves are vascularized by a dense network of blood vessels to guarantee their complex function. Despite the crucial role of vascularization to ensure nerve homeostasis and regeneration, the mechanisms governing nerve invasion by blood vessels remain poorly understood. We found, in mice, that the sciatic nerve invasion by blood vessels begins around embryonic day 16 and continues until birth. Interestingly, intra-nervous blood vessel density significantly decreases during post-natal period, starting from P10. We show that, while the axon guidance molecule Netrin-1 promotes nerve invasion by blood vessels via the endothelial receptor UNC5B during embryogenesis, myelinated Schwann cells negatively control intra-nervous vascularization during post-natal period.
Collapse
Affiliation(s)
- Sonia Taïb
- Center for Interdisciplinary Research in Biology, Collège de France, Paris, France
| | - Noël Lamandé
- Center for Interdisciplinary Research in Biology, Collège de France, Paris, France
| | - Sabrina Martin
- Center for Interdisciplinary Research in Biology, Collège de France, Paris, France
| | - Fanny Coulpier
- UMR U955 INSERM UPEC, Institut Mondor de Recherche Biomédicale, Créteil, France
| | - Piotr Topilko
- UMR U955 INSERM UPEC, Institut Mondor de Recherche Biomédicale, Créteil, France
| | - Isabelle Brunet
- Center for Interdisciplinary Research in Biology, Collège de France, Paris, France
| |
Collapse
|
20
|
Fralish Z, Lotz EM, Chavez T, Khodabukus A, Bursac N. Neuromuscular Development and Disease: Learning From in vitro and in vivo Models. Front Cell Dev Biol 2021; 9:764732. [PMID: 34778273 PMCID: PMC8579029 DOI: 10.3389/fcell.2021.764732] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Accepted: 10/06/2021] [Indexed: 01/02/2023] Open
Abstract
The neuromuscular junction (NMJ) is a specialized cholinergic synaptic interface between a motor neuron and a skeletal muscle fiber that translates presynaptic electrical impulses into motor function. NMJ formation and maintenance require tightly regulated signaling and cellular communication among motor neurons, myogenic cells, and Schwann cells. Neuromuscular diseases (NMDs) can result in loss of NMJ function and motor input leading to paralysis or even death. Although small animal models have been instrumental in advancing our understanding of the NMJ structure and function, the complexities of studying this multi-tissue system in vivo and poor clinical outcomes of candidate therapies developed in small animal models has driven the need for in vitro models of functional human NMJ to complement animal studies. In this review, we discuss prevailing models of NMDs and highlight the current progress and ongoing challenges in developing human iPSC-derived (hiPSC) 3D cell culture models of functional NMJs. We first review in vivo development of motor neurons, skeletal muscle, Schwann cells, and the NMJ alongside current methods for directing the differentiation of relevant cell types from hiPSCs. We further compare the efficacy of modeling NMDs in animals and human cell culture systems in the context of five NMDs: amyotrophic lateral sclerosis, myasthenia gravis, Duchenne muscular dystrophy, myotonic dystrophy, and Pompe disease. Finally, we discuss further work necessary for hiPSC-derived NMJ models to function as effective personalized NMD platforms.
Collapse
Affiliation(s)
| | | | | | | | - Nenad Bursac
- Department of Biomedical Engineering, Pratt School of Engineering, Duke University, Durham, NC, United States
| |
Collapse
|
21
|
Follis RM, Tep C, Genaro-Mattos TC, Kim ML, Ryu JC, Morrison VE, Chan JR, Porter N, Carter BD, Yoon SO. Metabolic Control of Sensory Neuron Survival by the p75 Neurotrophin Receptor in Schwann Cells. J Neurosci 2021; 41:8710-8724. [PMID: 34507952 PMCID: PMC8528492 DOI: 10.1523/jneurosci.3243-20.2021] [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: 12/31/2020] [Revised: 08/28/2021] [Accepted: 08/30/2021] [Indexed: 11/21/2022] Open
Abstract
We report that the neurotrophin receptor p75 contributes to sensory neuron survival through the regulation of cholesterol metabolism in Schwann cells. Selective deletion of p75 in mouse Schwann cells of either sex resulted in a 30% loss of dorsal root ganglia (DRG) neurons and diminished thermal sensitivity. P75 regulates Schwann cell cholesterol biosynthesis in response to BDNF, forming a co-receptor complex with ErbB2 and activating ErbB2-mediated stimulation of sterol regulatory element binding protein 2 (SREBP2), a master regulator of cholesterol synthesis. Schwann cells lacking p75 exhibited decreased activation of SREBP2 and a reduction in 7-dehydrocholesterol (7-DHC) reductase (DHCR7) expression, resulting in accumulation of the neurotoxic intermediate, 7-dehyrocholesterol in the sciatic nerve. Restoration of DHCR7 in p75 null Schwann cells in mice significantly attenuated DRG neuron loss. Together, these results reveal a mechanism by which the disruption of lipid metabolism in glial cells negatively influences sensory neuron survival, which has implications for a wide range of peripheral neuropathies.SIGNIFICANCE STATEMENT Although expressed in Schwann cells, the role of p75 in myelination has remained unresolved in part because of its dual expression in sensory neurons that Schwann cells myelinate. When p75 was deleted selectively among Schwann cells, myelination was minimally affected, while sensory neuron survival was reduced by 30%. The phenotype is mainly due to dysregulation of cholesterol biosynthesis in p75-deficient Schwann cells, leading to an accumulation of neurotoxic cholesterol precursor, 7-dehydrocholesterol (7-DHC). Mechanism-wise, we discovered that in response to BDNF, p75 recruits and activates ErbB2 independently of ErbB3, thereby stimulating the master regulator, sterol regulatory element binding protein 2 (SREBP2). These results together highlight a novel role of p75 in Schwann cells in regulating DRG neuron survival by orchestrating proper cholesterol metabolism.
Collapse
Affiliation(s)
- Rose M Follis
- Department of Biochemistry, Vanderbilt Brain Institute, Vanderbilt University School of Medicine, Nashville, Tennessee 37232
| | - Chhavy Tep
- Department of Biological Chemistry and Pharmacology, Ohio State University, Columbus, Ohio
| | - Thiago C Genaro-Mattos
- Department of Chemistry, Vanderbilt University School of Arts and Sciences, Nashville, Tennessee 37232
| | - Mi Lyang Kim
- Department of Biological Chemistry and Pharmacology, Ohio State University, Columbus, Ohio
| | - Jae Cheon Ryu
- Department of Biological Chemistry and Pharmacology, Ohio State University, Columbus, Ohio
| | - Vivianne E Morrison
- Department of Biochemistry, Vanderbilt Brain Institute, Vanderbilt University School of Medicine, Nashville, Tennessee 37232
| | - Jonah R Chan
- Department of Neurology, University of California San Francisco, San Francisco, California 94158
| | - Ned Porter
- Department of Chemistry, Vanderbilt University School of Arts and Sciences, Nashville, Tennessee 37232
| | - Bruce D Carter
- Department of Biochemistry, Vanderbilt Brain Institute, Vanderbilt University School of Medicine, Nashville, Tennessee 37232
| | - Sung Ok Yoon
- Department of Biological Chemistry and Pharmacology, Ohio State University, Columbus, Ohio
| |
Collapse
|
22
|
Kochat V, Raman AT, Landers SM, Tang M, Schulz J, Terranova C, Landry JP, Bhalla AD, Beird HC, Wu CC, Jiang Y, Mao X, Lazcano R, Gite S, Ingram DR, Yi M, Zhang J, Keung EZ, Scally CP, Roland CL, Hunt KK, Feig BW, Futreal PA, Hwu P, Wang WL, Lazar AJ, Slopis JM, Wilson-Robles H, Wiener DJ, McCutcheon IE, Wustefeld-Janssens B, Rai K, Torres KE. Enhancer reprogramming in PRC2-deficient malignant peripheral nerve sheath tumors induces a targetable de-differentiated state. Acta Neuropathol 2021; 142:565-590. [PMID: 34283254 DOI: 10.1007/s00401-021-02341-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Revised: 06/09/2021] [Accepted: 06/22/2021] [Indexed: 02/03/2023]
Abstract
Malignant peripheral nerve sheath tumors (MPNSTs) are soft tissue sarcomas that frequently harbor genetic alterations in polycomb repressor complex 2 (PRC2) components-SUZ12 and EED. Here, we show that PRC2 loss confers a dedifferentiated early neural-crest phenotype which is exclusive to PRC2-mutant MPNSTs and not a feature of neurofibromas. Neural crest phenotype in PRC2 mutant MPNSTs was validated via cross-species comparative analysis using spontaneous and transgenic MPNST models. Systematic chromatin state profiling of the MPNST cells showed extensive epigenomic reprogramming or chromatin states associated with PRC2 loss and identified gains of active enhancer states/super-enhancers on early neural crest regulators in PRC2-mutant conditions around genomic loci that harbored repressed/poised states in PRC2-WT MPNST cells. Consistently, inverse correlation between H3K27me3 loss and H3K27Ac gain was noted in MPNSTs. Epigenetic editing experiments established functional roles for enhancer gains on DLX5-a key regulator of neural crest phenotype. Consistently, blockade of enhancer activity by bromodomain inhibitors specifically suppressed this neural crest phenotype and tumor burden in PRC2-mutant PDXs. Together, these findings reveal accumulation of dedifferentiated neural crest like state in PRC2-mutant MPNSTs that can be targeted by enhancer blockade.
Collapse
Affiliation(s)
- Veena Kochat
- Department of Surgical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA.,Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Ayush T Raman
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.,Graduate Program in Quantitative and Computational Biosciences, Baylor College of Medicine, Houston, TX, USA.,Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Sharon M Landers
- Department of Surgical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Ming Tang
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.,Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Jonathan Schulz
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Christopher Terranova
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jace P Landry
- Department of Surgical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA.,Louisiana State University School of Medicine, New Orleans, LA, USA
| | - Angela D Bhalla
- Department of Surgical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Hannah C Beird
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Chia-Chin Wu
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Yingda Jiang
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.,Graduate School of Biomedical Sciences, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Xizeng Mao
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Rossana Lazcano
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Swati Gite
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Davis R Ingram
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Min Yi
- Department of Surgical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jianhua Zhang
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Emily Z Keung
- Department of Surgical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Christopher P Scally
- Department of Surgical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Christina L Roland
- Department of Surgical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Kelly K Hunt
- Department of Surgical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Barry W Feig
- Department of Surgical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - P Andrew Futreal
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Patrick Hwu
- Department of Melanoma Medical Oncology and Sarcoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.,H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Wei-Lien Wang
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Alexander J Lazar
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - John M Slopis
- Department of Neuro-Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Heather Wilson-Robles
- Small Animal Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX, USA
| | - Dominique J Wiener
- Department of Veterinary Pathobiology, College of Veterinary Medicine & Biomedical Science, Texas A&M University, College Station, TX, USA
| | - Ian E McCutcheon
- Department of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Brandan Wustefeld-Janssens
- Small Animal Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX, USA.,Department of Surgical Oncology, Flint Animal Cancer Center, Colorado State University, Fort Collins, CO, USA
| | - Kunal Rai
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA. .,Graduate Program in Quantitative and Computational Biosciences, Baylor College of Medicine, Houston, TX, USA.
| | - Keila E Torres
- Department of Surgical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA. .,Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA. .,Graduate School of Biomedical Sciences, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
23
|
Ishigami D, Miyawaki S, Nakatomi H, Takayanagi S, Teranishi Y, Ohara K, Hongo H, Dofuku S, Kin T, Abe H, Mitsui J, Komura D, Katoh H, Ishikawa S, Saito N. Brainstem intraparenchymal schwannoma with genetic analysis: a case report and literature review. BMC Med Genomics 2021; 14:205. [PMID: 34407809 PMCID: PMC8371869 DOI: 10.1186/s12920-021-01049-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2021] [Accepted: 07/28/2021] [Indexed: 12/02/2022] Open
Abstract
BACKGROUND Schwannomas are neoplasms that typically arise from the myelin sheath of peripheral nerves and rarely originate within the brain parenchyma. Some case reports present schwannomas arising from the brainstem, but regrowth of the tumor and the efficacy of postoperative irradiation have not been examined. In addition, the genetic background of schwannomas arising from the brainstem has not been investigated. CASE PRESENTATION A 21-year-old male presented with diplopia, dysphagia, and left-sided hemiparesis, dysesthesia, and ataxia. Intracranial imaging showed a heterogeneous mass with a cystic lesion in the pontomedullary junction. Since the tumor caused obstructive hydrocephalus, the patient underwent subtotal tumor resection. A histopathologic evaluation aided a diagnosis of brainstem intraparenchymal schwannoma. Gradual postoperative mass regrowth was recognized. Three-dimensional conformal radiotherapy was performed on the residual mass and surgical cavity. No tumor regrowth was observed 4 years after surgery. To investigate the genetic background of the tumor, target sequences for 36 genes, including NF2, SMARCB1, and LZTR1, and microsatellite analysis for loss of 22q did not show any somatic variants or 22q loss. CONCLUSIONS We suggest that brainstem schwannomas might differ from conventional schwannomas in their genetic background.
Collapse
Affiliation(s)
- Daiichiro Ishigami
- Department of Neurosurgery, Faculty of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Satoru Miyawaki
- Department of Neurosurgery, Faculty of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Hirofumi Nakatomi
- Department of Neurosurgery, Faculty of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan.
| | - Shunsaku Takayanagi
- Department of Neurosurgery, Faculty of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Yu Teranishi
- Department of Neurosurgery, Faculty of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Kenta Ohara
- Department of Neurosurgery, Faculty of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Hiroki Hongo
- Department of Neurosurgery, Faculty of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Shogo Dofuku
- Department of Neurosurgery, Faculty of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Taichi Kin
- Department of Neurosurgery, Faculty of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Hiroyuki Abe
- Department of Pathology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Jun Mitsui
- Department of Molecular Neurology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Daisuke Komura
- Department of Preventive Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Hiroto Katoh
- Department of Preventive Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Shumpei Ishikawa
- Department of Preventive Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Nobuhito Saito
- Department of Neurosurgery, Faculty of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| |
Collapse
|
24
|
Korkmaz Y, Imhof T, Kämmerer PW, Bloch W, Rink-Notzon S, Möst T, Weber M, Kesting M, Galler KM, Deschner J. The colocalizations of pulp neural stem cells markers with dentin matrix protein-1, dentin sialoprotein and dentin phosphoprotein in human denticle (pulp stone) lining cells. Ann Anat 2021; 239:151815. [PMID: 34400302 DOI: 10.1016/j.aanat.2021.151815] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Revised: 07/01/2021] [Accepted: 08/02/2021] [Indexed: 12/14/2022]
Abstract
BACKGROUND The primary dentin, secondary dentin, and reactive tertiary dentin are formed by terminal differentiated odontoblasts, whereas atubular reparative tertiary dentin is formed by odontoblast-like cells. Odontoblast-like cells differentiate from pulpal stem cells, which express the neural stem cell markers nestin, S100β, Sox10, and P0. The denticle (pulp stone) is an unique mineralized extracellular matrix that frequently occurs in association with the neurovascular structures in the dental pulp. However, to date, the cellular origin of denticles in human dental pulp is unclear. In addition, the non-collagenous extracellular dentin matrix proteins dentin matrix protein 1 (DMP1), dentin sialoprotein (DSP), and dentin phosphoprotein (DPP) have been well characterized in the dentin matrix, whereas their role in the formation and mineralization of the denticle matrix remains to be clarified. METHODS To characterize the formation of denticle, healthy human third molars (n = 59) were completely sectioned and evaluated by HE staining in different layers at 720 µm intervals. From these samples, molars with (n = 5) and without denticles (n = 8) were selected. Using consecutive cryo-sections from a layer containing denticles of different sizes, we examined DMP1, DSP, and DPP in denticle lining cells and tested their co-localizations with the glial stem cell markers nestin, S100β, Sox10, and P0 by quantitative and double staining methods. RESULTS DMP1, DSP and DPP were found in odontoblasts, whereas denticle lining cells were positive only for DMP1 and DSP but not for DPP. Nestin was detected in both odontoblasts and denticle lining cells. S100β, Sox10, and P0 were co-localized with DMP1 and DSP in different subpopulations of denticle lining cells. CONCLUSIONS The co-localization of S100β, Sox10, and P0 with DMP1 and DSP in denticle lining cells suggest that denticle lining cells are originated from glial and/or endoneurial mesenchymal stem cells which are involved in biomineralization of denticle matrix by secretion of DMP1 and DSP. Since denticles are atubular compared to primary, secondary, reactionary tertiary dentin and denticle formed by odontoblasts, our results suggest that DPP could be one of the proteins involved in the complex regulation of dentinal tubule formation.
Collapse
Affiliation(s)
- Yüksel Korkmaz
- Department of Periodontology and Operative Dentistry, University Medical Center of the Johannes Gutenberg University, Mainz, Germany.
| | - Thomas Imhof
- Institute for Experimental Dental Research and Oral Musculoskeletal Biology, Center for Biochemistry, Medical Faculty, University of Cologne, Cologne, Germany
| | - Peer W Kämmerer
- Department of Oral, and Maxillofacial and Plastic Surgery, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Wilhelm Bloch
- Institute of Cardiovascular Research and Sport Medicine, Department of Molecular and Cellular Sport Medicine, German Sport University Cologne, Cologne, Germany
| | - Svenja Rink-Notzon
- Department of Prosthetic Dentistry, School of Dental and Oral Medicine, University of Cologne, Cologne, Germany; Department of Pediatrics and Adolescent Medicine, Experimental Neonatology, Center for Biochemistry, Faculty of Medicine, University of Cologne, Cologne, Germany
| | - Tobias Möst
- Department of Oral and Maxillofacial Surgery, University Hospital Erlangen, Friedrich Alexander University, Erlangen, Germany
| | - Manuel Weber
- Department of Oral and Maxillofacial Surgery, University Hospital Erlangen, Friedrich Alexander University, Erlangen, Germany
| | - Marco Kesting
- Department of Oral and Maxillofacial Surgery, University Hospital Erlangen, Friedrich Alexander University, Erlangen, Germany
| | - Kerstin M Galler
- Department of Conservative Dentistry and Periodontology, University Hospital Regensburg, Regensburg, Germany
| | - James Deschner
- Department of Periodontology and Operative Dentistry, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| |
Collapse
|
25
|
Farina AR, Cappabianca LA, Zelli V, Sebastiano M, Mackay AR. Mechanisms involved in selecting and maintaining neuroblastoma cancer stem cell populations, and perspectives for therapeutic targeting. World J Stem Cells 2021; 13:685-736. [PMID: 34367474 PMCID: PMC8316860 DOI: 10.4252/wjsc.v13.i7.685] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Revised: 03/09/2021] [Accepted: 04/14/2021] [Indexed: 02/06/2023] Open
Abstract
Pediatric neuroblastomas (NBs) are heterogeneous, aggressive, therapy-resistant embryonal tumours that originate from cells of neural crest (NC) origin and in particular neuroblasts committed to the sympathoadrenal progenitor cell lineage. Therapeutic resistance, post-therapeutic relapse and subsequent metastatic NB progression are driven primarily by cancer stem cell (CSC)-like subpopulations, which through their self-renewing capacity, intermittent and slow cell cycles, drug-resistant and reversibly adaptive plastic phenotypes, represent the most important obstacle to improving therapeutic outcomes in unfavourable NBs. In this review, dedicated to NB CSCs and the prospects for their therapeutic eradication, we initiate with brief descriptions of the unique transient vertebrate embryonic NC structure and salient molecular protagonists involved NC induction, specification, epithelial to mesenchymal transition and migratory behaviour, in order to familiarise the reader with the embryonic cellular and molecular origins and background to NB. We follow this by introducing NB and the potential NC-derived stem/progenitor cell origins of NBs, before providing a comprehensive review of the salient molecules, signalling pathways, mechanisms, tumour microenvironmental and therapeutic conditions involved in promoting, selecting and maintaining NB CSC subpopulations, and that underpin their therapy-resistant, self-renewing metastatic behaviour. Finally, we review potential therapeutic strategies and future prospects for targeting and eradication of these bastions of NB therapeutic resistance, post-therapeutic relapse and metastatic progression.
Collapse
Affiliation(s)
- Antonietta Rosella Farina
- Department of Applied Clinical and Biotechnological Sciences, University of L'Aquila, L'Aquila 67100, AQ, Italy
| | - Lucia Annamaria Cappabianca
- Department of Applied Clinical and Biotechnological Sciences, University of L'Aquila, L'Aquila 67100, AQ, Italy
| | - Veronica Zelli
- Department of Applied Clinical and Biotechnological Sciences, University of L'Aquila, L'Aquila 67100, AQ, Italy
| | - Michela Sebastiano
- Department of Applied Clinical and Biotechnological Sciences, University of L'Aquila, L'Aquila 67100, AQ, Italy
| | - Andrew Reay Mackay
- Department of Applied Clinical and Biotechnological Sciences, University of L'Aquila, L'Aquila 67100, AQ, Italy.
| |
Collapse
|
26
|
Malavasi EL, Ghosh A, Booth DG, Zagnoni M, Sherman DL, Brophy PJ. Dynamic early clusters of nodal proteins contribute to node of Ranvier assembly during myelination of peripheral neurons. eLife 2021; 10:68089. [PMID: 34240706 PMCID: PMC8289411 DOI: 10.7554/elife.68089] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Accepted: 07/07/2021] [Indexed: 12/31/2022] Open
Abstract
Voltage-gated sodium channels cluster in macromolecular complexes at nodes of Ranvier to promote rapid nerve impulse conduction in vertebrate nerves. Node assembly in peripheral nerves is thought to be initiated at heminodes at the extremities of myelinating Schwann cells, and fusion of heminodes results in the establishment of nodes. Here we show that assembly of 'early clusters' of nodal proteins in the murine axonal membrane precedes heminode formation. The neurofascin (Nfasc) proteins are essential for node assembly, and the formation of early clusters also requires neuronal Nfasc. Early clusters are mobile and their proteins are dynamically recruited by lateral diffusion. They can undergo fusion not only with each other but also with heminodes, thus contributing to the development of nodes in peripheral axons. The formation of early clusters constitutes the earliest stage in peripheral node assembly and expands the repertoire of strategies that have evolved to establish these essential structures.
Collapse
Affiliation(s)
- Elise Lv Malavasi
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Aniket Ghosh
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Daniel G Booth
- Biodiscovery Institute, University of Nottingham, Nottingham, United Kingdom
| | - Michele Zagnoni
- Centre for Microsystems & Photonics, Dept. Electronic and Electrical Engineering, University of Strathclyde, Strathclyde, United Kingdom
| | - Diane L Sherman
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Peter J Brophy
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, United Kingdom
| |
Collapse
|
27
|
Echternacht SR, Chacon MA, Leckenby JI. Central versus peripheral nervous system regeneration: is there an exception for cranial nerves? Regen Med 2021; 16:567-579. [PMID: 34075805 DOI: 10.2217/rme-2020-0096] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
There exists a dichotomy in regenerative capacity between the PNS and CNS, which poses the question - where do cranial nerves fall? Through the discussion of the various cells and processes involved in axonal regeneration, we will evaluate whether the assumption that cranial nerve regeneration is analogous to peripheral nerve regeneration is valid. It is evident from this review that much remains to be clarified regarding both PNS and CNS regeneration. Furthermore, it is not clear if cranial nerves follow the PNS model, CNS model or possess an alternative novel regenerative process altogether. Future research should continue to focus on elucidating how cranial nerves regenerate; and the various cellular interactions, molecules and pathways involved.
Collapse
Affiliation(s)
- Scott R Echternacht
- University of Rochester School of Medicine & Dentistry, 601 Elmwood Avenue, Rochester, NY 14642, USA.,Division of Plastic Surgery, University of Rochester Medical Center, 601 Elmwood Avenue, Box 661, Rochester, NY 14642, USA
| | - Miranda A Chacon
- Division of Plastic Surgery, University of Rochester Medical Center, 601 Elmwood Avenue, Box 661, Rochester, NY 14642, USA.,Department of Surgery, University of Rochester Medical Center, 601 Elmwood Avenue, Box 661, Rochester, NY 14642, USA
| | - Jonathan I Leckenby
- Division of Plastic Surgery, University of Rochester Medical Center, 601 Elmwood Avenue, Box 661, Rochester, NY 14642, USA
| |
Collapse
|
28
|
Sundaram VK, El Jalkh T, Barakat R, Fernandez CJI, Massaad C, Grenier J. Retracing Schwann Cell Developmental Transitions in Embryonic Dissociated DRG/Schwann Cell Cocultures in Mice. Front Cell Neurosci 2021; 15:590537. [PMID: 34093128 PMCID: PMC8173108 DOI: 10.3389/fncel.2021.590537] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2020] [Accepted: 04/20/2021] [Indexed: 12/11/2022] Open
Abstract
Embryonic Dissociated Dorsal Root Ganglia (DRG) cultures are often used to investigate the role of novel molecular pathways or drugs in Schwann cell development and myelination. These cultures largely recapitulate the order of cellular and molecular events that occur in Schwann cells of embryonic nerves. However, the timing of Schwann cell developmental transitions, notably the transition from Schwann Cell Precursors (SCP) to immature Schwann cells (iSC) and then to myelinating Schwann cells, has not been estimated so far in this culture system. In this study, we determined the expression profiles of Schwann cell developmental genes during the first week of culture and then compared our data to the expression profiles of these genes in developing spinal nerves. This helped in identifying that SCP transition into iSC between the 5th and 7th day in vitro. Furthermore, we also investigated the transition of immature cells into pro-myelinating and myelinating Schwann cells upon the induction of myelination in vitro. Our results suggest that Schwann cell differentiation beyond the immature stage can be observed as early as 4 days post the induction of myelination in cocultures. Finally, we compared the myelinating potential of coculture-derived Schwann cell monocultures to cultures established from neonatal sciatic nerves and found that both these culture systems exhibit similar myelinating phenotypes. In effect, our results allow for a better understanding and interpretation of coculture experiments especially in studies that aim to elucidate the role of a novel actor in Schwann cell development and myelination.
Collapse
Affiliation(s)
| | - Tatiana El Jalkh
- INSERM UMRS 1124, Faculty of Basic and Biomedical Sciences, Université de Paris, Paris, France.,EC2M, Faculty of Sciences II, Lebanese University, Fanar, Lebanon
| | - Rasha Barakat
- INSERM UMRS 1124, Faculty of Basic and Biomedical Sciences, Université de Paris, Paris, France.,INSERM UMRS 1016, Institut Cochin, Université de Paris, Paris, France
| | | | - Charbel Massaad
- INSERM UMRS 1124, Faculty of Basic and Biomedical Sciences, Université de Paris, Paris, France
| | - Julien Grenier
- INSERM UMRS 1124, Faculty of Basic and Biomedical Sciences, Université de Paris, Paris, France
| |
Collapse
|
29
|
Nazareth L, St John J, Murtaza M, Ekberg J. Phagocytosis by Peripheral Glia: Importance for Nervous System Functions and Implications in Injury and Disease. Front Cell Dev Biol 2021; 9:660259. [PMID: 33898462 PMCID: PMC8060502 DOI: 10.3389/fcell.2021.660259] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Accepted: 03/17/2021] [Indexed: 12/30/2022] Open
Abstract
The central nervous system (CNS) has very limited capacity to regenerate after traumatic injury or disease. In contrast, the peripheral nervous system (PNS) has far greater capacity for regeneration. This difference can be partly attributed to variances in glial-mediated functions, such as axon guidance, structural support, secretion of growth factors and phagocytic activity. Due to their growth-promoting characteristic, transplantation of PNS glia has been trialed for neural repair. After peripheral nerve injuries, Schwann cells (SCs, the main PNS glia) phagocytose myelin debris and attract macrophages to the injury site to aid in debris clearance. One peripheral nerve, the olfactory nerve, is unique in that it continuously regenerates throughout life. The olfactory nerve glia, olfactory ensheathing cells (OECs), are the primary phagocytes within this nerve, continuously clearing axonal debris arising from the normal regeneration of the nerve and after injury. In contrast to SCs, OECs do not appear to attract macrophages. SCs and OECs also respond to and phagocytose bacteria, a function likely critical for tackling microbial invasion of the CNS via peripheral nerves. However, phagocytosis is not always effective; inflammation, aging and/or genetic factors may contribute to compromised phagocytic activity. Here, we highlight the diverse roles of SCs and OECs with the focus on their phagocytic activity under physiological and pathological conditions. We also explore why understanding the contribution of peripheral glia phagocytosis may provide us with translational strategies for achieving axonal regeneration of the injured nervous system and potentially for the treatment of certain neurological diseases.
Collapse
Affiliation(s)
- Lynn Nazareth
- Menzies Health Institute Queensland, Griffith University, Southport, QLD, Australia.,Clem Jones Centre for Neurobiology and Stem Cell Research, Griffith University, Nathan, QLD, Australia
| | - James St John
- Menzies Health Institute Queensland, Griffith University, Southport, QLD, Australia.,Clem Jones Centre for Neurobiology and Stem Cell Research, Griffith University, Nathan, QLD, Australia.,Griffith Institute for Drug Discovery, Griffith University, Nathan, QLD, Australia
| | - Mariyam Murtaza
- Menzies Health Institute Queensland, Griffith University, Southport, QLD, Australia.,Clem Jones Centre for Neurobiology and Stem Cell Research, Griffith University, Nathan, QLD, Australia.,Griffith Institute for Drug Discovery, Griffith University, Nathan, QLD, Australia
| | - Jenny Ekberg
- Menzies Health Institute Queensland, Griffith University, Southport, QLD, Australia.,Clem Jones Centre for Neurobiology and Stem Cell Research, Griffith University, Nathan, QLD, Australia.,Griffith Institute for Drug Discovery, Griffith University, Nathan, QLD, Australia
| |
Collapse
|
30
|
Vitamin E Deficiency Disrupts Gene Expression Networks during Zebrafish Development. Nutrients 2021; 13:nu13020468. [PMID: 33573233 PMCID: PMC7912379 DOI: 10.3390/nu13020468] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 01/26/2021] [Accepted: 01/27/2021] [Indexed: 02/08/2023] Open
Abstract
Vitamin E (VitE) is essential for vertebrate embryogenesis, but the mechanisms involved remain unknown. To study embryonic development, we fed zebrafish adults (>55 days) either VitE sufficient (E+) or deficient (E–) diets for >80 days, then the fish were spawned to generate E+ and E– embryos. To evaluate the transcriptional basis of the metabolic and phenotypic outcomes, E+ and E– embryos at 12, 18 and 24 h post-fertilization (hpf) were subjected to gene expression profiling by RNASeq. Hierarchical clustering, over-representation analyses and gene set enrichment analyses were performed with differentially expressed genes. E– embryos experienced overall disruption to gene expression associated with gene transcription, carbohydrate and energy metabolism, intracellular signaling and the formation of embryonic structures. mTOR was apparently a major controller of these changes. Thus, embryonic VitE deficiency results in genetic and transcriptional dysregulation as early as 12 hpf, leading to metabolic dysfunction and ultimately lethal outcomes.
Collapse
|
31
|
Feltri ML, Weaver MR, Belin S, Poitelon Y. The Hippo pathway: Horizons for innovative treatments of peripheral nerve diseases. J Peripher Nerv Syst 2021; 26:4-16. [PMID: 33449435 DOI: 10.1111/jns.12431] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Revised: 12/16/2020] [Accepted: 12/20/2020] [Indexed: 12/19/2022]
Abstract
Initially identified in Drosophila, the Hippo signaling pathway regulates how cells respond to their environment by controlling proliferation, migration and differentiation. Many recent studies have focused on characterizing Hippo pathway function and regulation in mammalian cells. Here, we present a brief overview of the major components of the Hippo pathway, as well as their regulation and function. We comprehensively review the studies that have contributed to our understanding of the Hippo pathway in the function of the peripheral nervous system and in peripheral nerve diseases. Finally, we discuss innovative approaches that aim to modulate Hippo pathway components in diseases of the peripheral nervous system.
Collapse
Affiliation(s)
- M Laura Feltri
- Hunter James Kelly Research Institute, Department of Biochemistry, Jacobs School of Medicine and Biomedical Sciences, Buffalo, New York, USA.,Department of Neurology, Jacobs School of Medicine and Biomedical Sciences, Buffalo, New York, USA
| | - Michael R Weaver
- Hunter James Kelly Research Institute, Department of Biochemistry, Jacobs School of Medicine and Biomedical Sciences, Buffalo, New York, USA
| | - Sophie Belin
- Department of Neuroscience and Experimental Therapeutics, Albany Medical College, Albany, New York, USA
| | - Yannick Poitelon
- Department of Neuroscience and Experimental Therapeutics, Albany Medical College, Albany, New York, USA
| |
Collapse
|
32
|
Muppirala AN, Limbach LE, Bradford EF, Petersen SC. Schwann cell development: From neural crest to myelin sheath. WILEY INTERDISCIPLINARY REVIEWS-DEVELOPMENTAL BIOLOGY 2020; 10:e398. [PMID: 33145925 DOI: 10.1002/wdev.398] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Revised: 10/06/2020] [Accepted: 10/07/2020] [Indexed: 12/16/2022]
Abstract
Vertebrate nervous system function requires glial cells, including myelinating glia that insulate axons and provide trophic support that allows for efficient signal propagation by neurons. In vertebrate peripheral nervous systems, neural crest-derived glial cells known as Schwann cells (SCs) generate myelin by encompassing and iteratively wrapping membrane around single axon segments. SC gliogenesis and neurogenesis are intimately linked and governed by a complex molecular environment that shapes their developmental trajectory. Changes in this external milieu drive developing SCs through a series of distinct morphological and transcriptional stages from the neural crest to a variety of glial derivatives, including the myelinating sublineage. Cues originate from the extracellular matrix, adjacent axons, and the developing SC basal lamina to trigger intracellular signaling cascades and gene expression changes that specify stages and transitions in SC development. Here, we integrate the findings from in vitro neuron-glia co-culture experiments with in vivo studies investigating SC development, particularly in zebrafish and mouse, to highlight critical factors that specify SC fate. Ultimately, we connect classic biochemical and mutant studies with modern genetic and visualization tools that have elucidated the dynamics of SC development. This article is categorized under: Signaling Pathways > Cell Fate Signaling Nervous System Development > Vertebrates: Regional Development.
Collapse
Affiliation(s)
- Anoohya N Muppirala
- Program in Neuroscience, Harvard Medical School, Boston, Massachusetts, USA.,Department of Neuroscience, Kenyon College, Gambier, Ohio, USA
| | | | | | - Sarah C Petersen
- Department of Neuroscience, Kenyon College, Gambier, Ohio, USA.,Department of Biology, Kenyon College, Gambier, Ohio, USA
| |
Collapse
|
33
|
Zhang R, Chen S, Wang X, Gu X, Yi S. Cell populations in neonatal rat peripheral nerves identified by single-cell transcriptomics. Glia 2020; 69:765-778. [PMID: 33079428 DOI: 10.1002/glia.23928] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2020] [Revised: 10/07/2020] [Accepted: 10/08/2020] [Indexed: 12/18/2022]
Abstract
Peripheral nerves connect central nerves with target tissues and organs and execute vital signal transduction functions. Although sub-types of neurons have been defined, the heterogeneity of cell populations in peripheral nerves, especially Schwann cells, has not been well demonstrated. Here, we collected sciatic nerves (SN) and dorsal root ganglia (DRG) from neonatal (1-day old) rats and classified cell populations by high-coverage single-cell sequencing. A total of 10 types of cells, including endothelial cells, erythrocytes, fibroblasts, monocytic cells, neurons, neutrophils, pericytes, satellite cells, Schwann cells, and vascular smooth muscle cells, were identified by transcriptome-based cell typing. The comparisons of cells in neonatal rat SN and DRG revealed distinct atlas in different tissue localizations. Investigations of ligand-receptor interactions showed that there existed direct cell-cell communications between endothelial cells and fibroblasts in SN and among endothelial cells, fibroblasts, and vascular smooth muscle cells in DRG. Schwann cells in neonatal rats were further sub-grouped to four sub-types, including LOC100134871 and Hbb expressing Schwann cell sub-type 1, Cldn19 and Emid1 expressing Schwann cell sub-type 2, Timp3 and Col5a3 expressing Schwann cell sub-type 3, and Cenpf and Mki67 expressing Schwann cell sub-type 4. These Schwann cell sub-types exhibited distinct genetic features and functional enrichments. Collectively, our results illustrated the diversity and cellular complexity of peripheral nerves at the neonatal stage and revealed the heterogeneity of Schwann cells in the peripheral nervous system.
Collapse
Affiliation(s)
- Ruirui Zhang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu, China
| | - Sailing Chen
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu, China
| | - Xinghui Wang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu, China
| | - Xiaosong Gu
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu, China
| | - Sheng Yi
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu, China
| |
Collapse
|
34
|
Yoon J, Barton MJ, St John JA, Ekberg JAK, Khan A, Redmond M. Anterior skull base olfactory tumours, which is what? A case report and review. J Clin Neurosci 2020; 81:1-5. [PMID: 33222894 DOI: 10.1016/j.jocn.2020.09.008] [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: 12/26/2019] [Revised: 07/29/2020] [Accepted: 09/06/2020] [Indexed: 11/17/2022]
Abstract
Intracranial schwannomas not originating from cranial nerves are rare. In this paper, we report a case of a 50-year-old male who presented with worsening headaches, diplopia and nausea over two years. Radiological imaging revealed a large tumour arising from the olfactory groove region with a preoperative diagnosis of olfactory groove meningioma (OGM). Intraoperatively, the tumour originated from the region of the attachment of the falx to the crista galli. The patient recovered without complication and histopathology reported an unexpected diagnosis of WHO Grade 1 schwannoma. However, as olfactory groove schwannomas (OGSs) cannot be distinguished from olfactory ensheathing cell tumours (OECTs), it is possible that the tumour could have been either an OGS or an OECT. Distinguishing between OGSs, OECTs and OGMs preoperatively is difficult. OGMs exhibit distinct histopathological features from OGSs/OECTs, however, OGSs and OECTs currently cannot be distinguished from each other. Here, we review the literature to discuss the differentiating features and cellular origins of these three tumours.
Collapse
Affiliation(s)
- J Yoon
- Neurosurgery Department, Royal Darwin Hospital, Darwin, Northern Territory, Australia
| | - M J Barton
- Menzies Health Institute Queensland, Griffith University, Gold Coast, Queensland, Australia; School of Nursing and Midwifery, Griffith University, Nathan, QLD, Australia
| | - J A St John
- Menzies Health Institute Queensland, Griffith University, Gold Coast, Queensland, Australia; Griffith Institute for Drug Discovery, Griffith University, Nathan, Queensland, Australia
| | - J A K Ekberg
- Menzies Health Institute Queensland, Griffith University, Gold Coast, Queensland, Australia; Griffith Institute for Drug Discovery, Griffith University, Nathan, Queensland, Australia
| | - A Khan
- Anatomical Pathology, Royal Darwin Hospital, Darwin, Northern Territory, Australia
| | - M Redmond
- Neurosurgery Department, Royal Darwin Hospital, Darwin, Northern Territory, Australia; Kenneth G Jamieson Department of Neurosurgery, Royal Brisbane and Women's Hospital, Brisbane, Queensland, Australia.
| |
Collapse
|
35
|
Kerman BE, Genoud S, Kurt Vatandaslar B, Denli AM, Georges Ghosh S, Xu X, Yeo GW, Aimone JB, Gage FH. Motoneuron expression profiling identifies an association between an axonal splice variant of HDGF-related protein 3 and peripheral myelination. J Biol Chem 2020; 295:12233-12246. [PMID: 32647008 PMCID: PMC7443494 DOI: 10.1074/jbc.ra120.014329] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Revised: 06/27/2020] [Indexed: 11/06/2022] Open
Abstract
Disorders that disrupt myelin formation during development or in adulthood, such as multiple sclerosis and peripheral neuropathies, lead to severe pathologies, illustrating myelin's crucial role in normal neural functioning. However, although our understanding of glial biology is increasing, the signals that emanate from axons and regulate myelination remain largely unknown. To identify the core components of the myelination process, here we adopted a microarray analysis approach combined with laser-capture microdissection of spinal motoneurons during the myelinogenic phase of development. We identified neuronal genes whose expression was enriched during myelination and further investigated hepatoma-derived growth factor-related protein 3 (HRP3 or HDGFRP3). HRP3 was strongly expressed in the white matter fiber tracts of the peripheral (PNS) and central (CNS) nervous systems during myelination and remyelination in a cuprizone-induced demyelination model. The dynamic localization of HPR3 between axons and nuclei during myelination was consistent with its axonal localization during neuritogenesis. To study this phenomenon, we identified two splice variants encoded by the HRP3 gene: the canonical isoform HRP3-I and a newly recognized isoform, HRP3-II. HRP3-I remained solely in the nucleus, whereas HRP3-II displayed distinct axonal localization both before and during myelination. Interestingly, HRP3-II remained in the nuclei of unmyelinated neurons and glial cells, suggesting the existence of a molecular machinery that transfers it to and retains it in the axons of neurons fated for myelination. Overexpression of HRP3-II, but not of HRP3-I, increased Schwann cell numbers and myelination in PNS neuron-glia co-cultures. However, HRP3-II overexpression in CNS co-cultures did not alter myelination.
Collapse
Affiliation(s)
- Bilal Ersen Kerman
- Department of Histology and Embryology, Istanbul Medipol University International School of Medicine, Istanbul, Turkey; Regenerative and Restorative Medicine Research Center, Institute of Health Science, Department of Neuroscience, Istanbul Medipol University, Istanbul, Turkey; Laboratory of Genetics, The Salk Institute for Biological Studies, La Jolla, California, USA
| | - Stéphane Genoud
- Laboratory of Genetics, The Salk Institute for Biological Studies, La Jolla, California, USA; Vifor Pharma, Villars-sur-Glâne, Switzerland
| | - Burcu Kurt Vatandaslar
- Regenerative and Restorative Medicine Research Center, Institute of Health Science, Department of Neuroscience, Istanbul Medipol University, Istanbul, Turkey; Institute of Health Science, Department of Neuroscience, Istanbul Medipol University, Istanbul, Turkey
| | - Ahmet Murat Denli
- Laboratory of Genetics, The Salk Institute for Biological Studies, La Jolla, California, USA
| | - Shereen Georges Ghosh
- Laboratory for Pediatric Brain Disease, University of California, San Diego, La Jolla, California, USA; Rady Children's Institute for Genomic Medicine, Rady Children's Hospital, San Diego, California, USA
| | - Xiangdong Xu
- Department of Pathology, University of California, San Diego, La Jolla, California, USA
| | - Gene W Yeo
- Department of Cellular and Molecular Medicine, Institute for Genomic Medicine, UCSD Stem Cell Program, University of California, San Diego, La Jolla, California, USA
| | - James Bradley Aimone
- Center for Computing Research, Sandia National Laboratories, Albuquerque, New Mexico, USA
| | - Fred H Gage
- Laboratory of Genetics, The Salk Institute for Biological Studies, La Jolla, California, USA.
| |
Collapse
|
36
|
Patchett AL, Flies AS, Lyons AB, Woods GM. Curse of the devil: molecular insights into the emergence of transmissible cancers in the Tasmanian devil (Sarcophilus harrisii). Cell Mol Life Sci 2020; 77:2507-2525. [PMID: 31900624 PMCID: PMC11104928 DOI: 10.1007/s00018-019-03435-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2019] [Revised: 12/17/2019] [Accepted: 12/19/2019] [Indexed: 12/22/2022]
Abstract
The Tasmanian devil (Sarcophilus harrisii) is the only mammalian species known to be affected by multiple transmissible cancers. Devil facial tumours 1 and 2 (DFT1 and DFT2) are independent neoplastic cell lineages that produce large, disfiguring cancers known as devil facial tumour disease (DFTD). The long-term persistence of wild Tasmanian devils is threatened due to the ability of DFTD cells to propagate as contagious allografts and the high mortality rate of DFTD. Recent studies have demonstrated that both DFT1 and DFT2 cancers originated from founder cells of the Schwann cell lineage, an uncommon origin of malignant cancer in humans. This unprecedented finding has revealed a potential predisposition of Tasmanian devils to transmissible cancers of the Schwann cell lineage. In this review, we compare the molecular nature of human Schwann cells and nerve sheath tumours with DFT1 and DFT2 to gain insights into the emergence of transmissible cancers in the Tasmanian devil. We discuss a potential mechanism, whereby Schwann cell plasticity and frequent wounding in Tasmanian devils combine with an inherent cancer predisposition and low genetic diversity to give rise to transmissible Schwann cell cancers in devils on rare occasions.
Collapse
Affiliation(s)
- Amanda L Patchett
- Menzies Institute for Medical Research, University of Tasmania, 17 Liverpool Street, Hobart, TAS, 7000, Australia
| | - Andrew S Flies
- Menzies Institute for Medical Research, University of Tasmania, 17 Liverpool Street, Hobart, TAS, 7000, Australia
| | - A Bruce Lyons
- School of Medicine, University of Tasmania, Hobart, TAS, 7000, Australia
| | - Gregory M Woods
- Menzies Institute for Medical Research, University of Tasmania, 17 Liverpool Street, Hobart, TAS, 7000, Australia.
| |
Collapse
|
37
|
Colazo JM, Evans BC, Farinas AF, Al-Kassis S, Duvall CL, Thayer WP. Applied Bioengineering in Tissue Reconstruction, Replacement, and Regeneration. TISSUE ENGINEERING PART B-REVIEWS 2020; 25:259-290. [PMID: 30896342 DOI: 10.1089/ten.teb.2018.0325] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
IMPACT STATEMENT The use of autologous tissue in the reconstruction of tissue defects has been the gold standard. However, current standards still face many limitations and complications. Improving patient outcomes and quality of life by addressing these barriers remain imperative. This article provides historical perspective, covers the major limitations of current standards of care, and reviews recent advances and future prospects in applied bioengineering in the context of tissue reconstruction, replacement, and regeneration.
Collapse
Affiliation(s)
- Juan M Colazo
- 1Vanderbilt University School of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee.,2Medical Scientist Training Program, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Brian C Evans
- 3Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee
| | - Angel F Farinas
- 4Department of Plastic Surgery, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Salam Al-Kassis
- 4Department of Plastic Surgery, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Craig L Duvall
- 3Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee
| | - Wesley P Thayer
- 3Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee.,4Department of Plastic Surgery, Vanderbilt University Medical Center, Nashville, Tennessee
| |
Collapse
|
38
|
Bouçanova F, Chrast R. Metabolic Interaction Between Schwann Cells and Axons Under Physiological and Disease Conditions. Front Cell Neurosci 2020; 14:148. [PMID: 32547370 PMCID: PMC7274022 DOI: 10.3389/fncel.2020.00148] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Accepted: 05/04/2020] [Indexed: 12/24/2022] Open
Abstract
Recent research into axon-glial interactions in the nervous system has made a compelling case that glial cells have a relevant role in the metabolic support of axons, and that, in the case of myelinating cells, this role is independent of myelination itself. In this mini-review article, we summarize some of those observations and focus on Schwann cells (SC), drawing parallels between glia of the central and peripheral nervous systems (PNS), pointing out limitations in current knowledge, and discussing its potential clinical relevance. First, we introduce SC, their development and main roles, and follow with an evolutionary perspective of glial metabolic function. Then we provide evidence of the myelin-independent aspects of axonal support and their coupling to neuronal metabolism. Finally, we address the opportunity to use SC-axon metabolic interactions as therapeutic targets to treat peripheral neuropathies.
Collapse
Affiliation(s)
- Filipa Bouçanova
- Department of Neuroscience, KarolinskaInstitutet, Stockholm, Sweden.,Department of Clinical Neuroscience, KarolinskaInstitutet, Stockholm, Sweden
| | - Roman Chrast
- Department of Neuroscience, KarolinskaInstitutet, Stockholm, Sweden.,Department of Clinical Neuroscience, KarolinskaInstitutet, Stockholm, Sweden
| |
Collapse
|
39
|
Zhang X, Murray B, Mo G, Shern JF. The Role of Polycomb Repressive Complex in Malignant Peripheral Nerve Sheath Tumor. Genes (Basel) 2020; 11:genes11030287. [PMID: 32182803 PMCID: PMC7140867 DOI: 10.3390/genes11030287] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Revised: 02/24/2020] [Accepted: 03/02/2020] [Indexed: 12/24/2022] Open
Abstract
Malignant peripheral nerve sheath tumors (MPNSTs) are aggressive soft tissue sarcomas that can arise most frequently in patients with neurofibromatosis type 1 (NF1). Despite an increasing understanding of the molecular mechanisms that underlie these tumors, there remains limited therapeutic options for this aggressive disease. One potentially critical finding is that a significant proportion of MPNSTs exhibit recurrent mutations in the genes EED or SUZ12, which are key components of the polycomb repressive complex 2 (PRC2). Tumors harboring these genetic lesions lose the marker of transcriptional repression, trimethylation of lysine residue 27 on histone H3 (H3K27me3) and have dysregulated oncogenic signaling. Given the recurrence of PRC2 alterations, intensive research efforts are now underway with a focus on detailing the epigenetic and transcriptomic consequences of PRC2 loss as well as development of novel therapeutic strategies for targeting these lesions. In this review article, we will summarize the recent findings of PRC2 in MPNST tumorigenesis, including highlighting the functions of PRC2 in normal Schwann cell development and nerve injury repair, as well as provide commentary on the potential therapeutic vulnerabilities of a PRC2 deficient tumor cell.
Collapse
Affiliation(s)
- Xiyuan Zhang
- Pediatric Oncology Branch, Tumor Evolution and Genomics Section, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA; (X.Z.); (B.M.); (G.M.)
| | - Béga Murray
- Pediatric Oncology Branch, Tumor Evolution and Genomics Section, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA; (X.Z.); (B.M.); (G.M.)
- The Patrick G Johnston Centre for Cancer Research, Queen’s University Belfast, 97 Lisburn road, Belfast BT9 7AE, UK
| | - George Mo
- Pediatric Oncology Branch, Tumor Evolution and Genomics Section, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA; (X.Z.); (B.M.); (G.M.)
- SUNY Downstate Health Sciences University, Brooklyn, NY 11203, USA
| | - Jack F. Shern
- Pediatric Oncology Branch, Tumor Evolution and Genomics Section, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA; (X.Z.); (B.M.); (G.M.)
- Correspondence:
| |
Collapse
|
40
|
Duman M, Martinez-Moreno M, Jacob C, Tapinos N. Functions of histone modifications and histone modifiers in Schwann cells. Glia 2020; 68:1584-1595. [PMID: 32034929 DOI: 10.1002/glia.23795] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Revised: 01/24/2020] [Accepted: 01/28/2020] [Indexed: 01/25/2023]
Abstract
Schwann cells (SCs) are the main glial cells present in the peripheral nervous system (PNS). Their primary functions are to insulate peripheral axons to protect them from the environment and to enable fast conduction of electric signals along big caliber axons by enwrapping them in a thick myelin sheath rich in lipids. In addition, SCs have the peculiar ability to foster axonal regrowth after a lesion by demyelinating and converting into repair cells that secrete neurotrophic factors and guide axons back to their former target to finally remyelinate regenerated axons. The different steps of SC development and their role in the maintenance of PNS integrity and regeneration after lesion are controlled by various factors among which transcription factors and chromatin-remodeling enzymes hold major functions. In this review, we discussed how histone modifications and histone-modifying enzymes control SC development, maintenance of PNS integrity and response to injury. The functions of histone modifiers as part of chromatin-remodeling complexes are discussed in another review published in the same issue of Glia.
Collapse
Affiliation(s)
- Mert Duman
- Faculty of Biology, Institute of Developmental Biology and Neurobiology, Johannes Gutenberg University Mainz, Mainz, Germany
| | - Margot Martinez-Moreno
- Department of Neurosurgery, Molecular Neuroscience & Neuro-Oncology Laboratory, Brown University, Providence, Rhode Island
| | - Claire Jacob
- Faculty of Biology, Institute of Developmental Biology and Neurobiology, Johannes Gutenberg University Mainz, Mainz, Germany
| | - Nikos Tapinos
- Department of Neurosurgery, Molecular Neuroscience & Neuro-Oncology Laboratory, Brown University, Providence, Rhode Island
| |
Collapse
|
41
|
Oligodendrocytes in Development, Myelin Generation and Beyond. Cells 2019; 8:cells8111424. [PMID: 31726662 PMCID: PMC6912544 DOI: 10.3390/cells8111424] [Citation(s) in RCA: 345] [Impact Index Per Article: 57.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Revised: 11/07/2019] [Accepted: 11/07/2019] [Indexed: 02/06/2023] Open
Abstract
Oligodendrocytes are the myelinating cells of the central nervous system (CNS) that are generated from oligodendrocyte progenitor cells (OPC). OPC are distributed throughout the CNS and represent a pool of migratory and proliferative adult progenitor cells that can differentiate into oligodendrocytes. The central function of oligodendrocytes is to generate myelin, which is an extended membrane from the cell that wraps tightly around axons. Due to this energy consuming process and the associated high metabolic turnover oligodendrocytes are vulnerable to cytotoxic and excitotoxic factors. Oligodendrocyte pathology is therefore evident in a range of disorders including multiple sclerosis, schizophrenia and Alzheimer’s disease. Deceased oligodendrocytes can be replenished from the adult OPC pool and lost myelin can be regenerated during remyelination, which can prevent axonal degeneration and can restore function. Cell population studies have recently identified novel immunomodulatory functions of oligodendrocytes, the implications of which, e.g., for diseases with primary oligodendrocyte pathology, are not yet clear. Here, we review the journey of oligodendrocytes from the embryonic stage to their role in homeostasis and their fate in disease. We will also discuss the most common models used to study oligodendrocytes and describe newly discovered functions of oligodendrocytes.
Collapse
|
42
|
Bradley EC, Cunningham RL, Wilde C, Morgan RK, Klug EA, Letcher SM, Schöneberg T, Monk KR, Liebscher I, Petersen SC. In vivo identification of small molecules mediating Gpr126/Adgrg6 signaling during Schwann cell development. Ann N Y Acad Sci 2019; 1456:44-63. [PMID: 31529518 PMCID: PMC7189964 DOI: 10.1111/nyas.14233] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Revised: 07/30/2019] [Accepted: 08/26/2019] [Indexed: 12/13/2022]
Abstract
Gpr126/Adgrg6, an adhesion family G protein-coupled receptor (aGPCR), is required for the development of myelinating Schwann cells in the peripheral nervous system. Myelin supports and insulates vertebrate axons to permit rapid signal propagation throughout the nervous system. In mammals and zebrafish, mutations in Gpr126 arrest Schwann cells at early developmental stages. We exploited the optical and pharmacological tractability of larval zebrafish to uncover drugs that mediate myelination by activating Gpr126 or functioning in parallel. Using a fluorescent marker of mature myelinating glia (Tg[mbp:EGFP-CAAX]), we screened hypomorphic gpr126 mutant larvae for restoration of myelin basic protein (mbp) expression along peripheral nerves following small molecule treatment. Our screens identified five compounds sufficient to promote mbp expression in gpr126 hypomorphs. Using an allelic series of gpr126 mutants, we parsed the ability of small molecules to restore mbp, suggesting differences in drug efficacy dependent on Schwann cell developmental state. Finally, we identify apomorphine hydrochloride as a direct small molecule activator of Gpr126 using combined in vivo/in vitro assays and show that aporphine class compounds promote Schwann cell development in vivo. Our results demonstrate the utility of in vivo screening for aGPCR modulators and identify small molecules that interact with the gpr126-mediated myelination program.
Collapse
Affiliation(s)
| | - Rebecca L. Cunningham
- Department of Developmental Biology, Washington University in St. Louis School of Medicine, St. Louis, MO, USA
| | - Caroline Wilde
- Rudolf Schönheimer Institute of Biochemistry, Medical Faculty, University of Leipzig, Leipzig, Germany
| | - Rory K. Morgan
- Vollum Institute, Oregon Health & Science University, Portland, OR, USA
| | - Emma A. Klug
- Department of Neuroscience, Kenyon College, Gambier, OH, USA
| | | | - Torsten Schöneberg
- Rudolf Schönheimer Institute of Biochemistry, Medical Faculty, University of Leipzig, Leipzig, Germany
| | - Kelly R. Monk
- Department of Developmental Biology, Washington University in St. Louis School of Medicine, St. Louis, MO, USA
- Vollum Institute, Oregon Health & Science University, Portland, OR, USA
| | - Ines Liebscher
- Rudolf Schönheimer Institute of Biochemistry, Medical Faculty, University of Leipzig, Leipzig, Germany
| | - Sarah C. Petersen
- Department of Neuroscience, Kenyon College, Gambier, OH, USA
- Department of Developmental Biology, Washington University in St. Louis School of Medicine, St. Louis, MO, USA
| |
Collapse
|
43
|
Sundaram VK, Massaad C, Grenier J. Liver X Receptors and Their Implications in the Physiology and Pathology of the Peripheral Nervous System. Int J Mol Sci 2019; 20:ijms20174192. [PMID: 31461876 PMCID: PMC6747127 DOI: 10.3390/ijms20174192] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Revised: 08/14/2019] [Accepted: 08/19/2019] [Indexed: 02/07/2023] Open
Abstract
Recent research in the last decade has sought to explore the role and therapeutic potential of Liver X Receptors (LXRs) in the physiology and pathologies of the Peripheral Nervous System. LXRs have been shown to be important in maintaining the redox homeostasis in peripheral nerves for proper myelination, and they regulate ER stress in sensory neurons. Furthermore, LXR stimulation has a positive impact on abrogating the effects of diabetic peripheral neuropathy and obesity-induced allodynia in the Peripheral Nervous System (PNS). This review details these findings and addresses certain important questions that are yet to be answered. The potential roles of LXRs in different cells of the PNS are speculated based on existing knowledge. The review also aims to provide important perspectives for further research in elucidating the role of LXRs and assessing the potential of LXR based therapies to combat pathologies of the Peripheral Nervous System.
Collapse
Affiliation(s)
- Venkat Krishnan Sundaram
- Faculty of Basic and Biomedical Sciences, Paris Descartes University, INSERM UMRS 1124, 75006 Paris, France
| | - Charbel Massaad
- Faculty of Basic and Biomedical Sciences, Paris Descartes University, INSERM UMRS 1124, 75006 Paris, France
| | - Julien Grenier
- Faculty of Basic and Biomedical Sciences, Paris Descartes University, INSERM UMRS 1124, 75006 Paris, France.
| |
Collapse
|
44
|
Liu T, Li G, Noble KV, Li Y, Barth JL, Schulte BA, Lang H. Age-dependent alterations of Kir4.1 expression in neural crest-derived cells of the mouse and human cochlea. Neurobiol Aging 2019; 80:210-222. [PMID: 31220650 PMCID: PMC6679794 DOI: 10.1016/j.neurobiolaging.2019.04.009] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2018] [Revised: 04/02/2019] [Accepted: 04/11/2019] [Indexed: 11/18/2022]
Abstract
Age-related hearing loss (or presbyacusis) is a progressive pathophysiological process. This study addressed the hypothesis that degeneration/dysfunction of multiple nonsensory cell types contributes to presbyacusis by evaluating tissues obtained from young and aged CBA/CaJ mouse ears and human temporal bones. Ultrastructural examination and transcriptomic analysis of mouse cochleas revealed age-dependent pathophysiological alterations in 3 types of neural crest-derived cells, namely intermediate cells in the stria vascularis, outer sulcus cells in the cochlear lateral wall, and satellite cells in the spiral ganglion. A significant decline in immunoreactivity for Kir4.1, an inwardly rectifying potassium channel, was seen in strial intermediate cells and outer sulcus cells in the ears of older mice. Age-dependent alterations in Kir4.1 immunostaining also were observed in satellite cells ensheathing spiral ganglion neurons. Expression alterations of Kir4.1 were observed in these same cell populations in the aged human cochlea. These results suggest that degeneration/dysfunction of neural crest-derived cells maybe an important contributing factor to both metabolic and neural forms of presbyacusis.
Collapse
Affiliation(s)
- Ting Liu
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, SC, USA; Department of Otolaryngology Head and Neck Surgery, Beijing Tongren Hospital, Capital Medical University, Key Laboratory of Otolaryngology Head and Neck Surgery, Ministry of Education, Beijing, China
| | - Gang Li
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, SC, USA; Department of Otolaryngology, Tinnitus and Hyperacusis Center, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Kenyaria V Noble
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, SC, USA
| | - Yongxi Li
- Department of Otolaryngology Head and Neck Surgery, Beijing Tongren Hospital, Capital Medical University, Key Laboratory of Otolaryngology Head and Neck Surgery, Ministry of Education, Beijing, China
| | - Jeremy L Barth
- Department of Regenerative Medicine, Medical University of South Carolina, Charleston, SC, USA
| | - Bradley A Schulte
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, SC, USA; Department of Otolaryngology-Head and Neck Surgery, Medical University of South Carolina, Charleston, SC, USA
| | - Hainan Lang
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, SC, USA.
| |
Collapse
|
45
|
Miranda IC, Taylor KR, Castleman W, de Lahunta A, Summers BA, Miller AD. Schwannosis in Three Foals and a Calf. Vet Pathol 2019; 56:783-788. [PMID: 31109258 DOI: 10.1177/0300985819846872] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Proliferation of ectopic Schwann cells within the central nervous system (CNS) parenchyma (schwannosis) in early life is most commonly associated with human neurofibromatosis type-2 and has been unrecognized in domestic animals. Three foals and a calf, 5 to 11 weeks old, with progressive neurological signs from birth were studied. Histologically, at multiple levels of the spinal cord, all animals had bilateral plaques of proliferative spindle cells, predominantly affecting the white matter adjacent to dorsal and ventral nerve roots and variably extending into the gray matter. Proliferating cells had strong intracytoplasmic immunoreactivity for the Schwann cell markers myelin protein zero and periaxin, highlighting the formation of peripheral nervous system (PNS) myelin within the spinal cord. In all cases, foci of disorganized neural tissue (glioneuronal hamartomas) were present, which in 2 cases formed a mass effect that resulted in syringohydromyelia. Neonatal presentation suggests a congenital maldevelopment of the nervous system, with spontaneous invasion of PNS-derived Schwann cells into the CNS.
Collapse
Affiliation(s)
- Ileana C Miranda
- 1 Department of Biomedical Sciences, Cornell University College of Veterinary Medicine, Section of Anatomic Pathology, Ithaca, NY, USA
| | - Kyle R Taylor
- 2 Washington Animal Disease Diagnostic Laboratory and Department of Veterinary Microbiology and Pathology, Washington State University College of Veterinary Medicine, Pullman, WA, USA
| | - William Castleman
- 3 University of Florida, College of Veterinary Medicine, Gainesville, FL, USA
| | - Alexander de Lahunta
- 1 Department of Biomedical Sciences, Cornell University College of Veterinary Medicine, Section of Anatomic Pathology, Ithaca, NY, USA
| | - Brian A Summers
- 1 Department of Biomedical Sciences, Cornell University College of Veterinary Medicine, Section of Anatomic Pathology, Ithaca, NY, USA
| | - Andrew D Miller
- 1 Department of Biomedical Sciences, Cornell University College of Veterinary Medicine, Section of Anatomic Pathology, Ithaca, NY, USA
| |
Collapse
|
46
|
Cheong H, Kim J, Kim BJ, Kim E, Park HY, Choi BH, Joo KI, Cho ML, Rhie JW, Lee JI, Cha HJ. Multi-dimensional bioinspired tactics using an engineered mussel protein glue-based nanofiber conduit for accelerated functional nerve regeneration. Acta Biomater 2019; 90:87-99. [PMID: 30978510 DOI: 10.1016/j.actbio.2019.04.018] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2018] [Revised: 04/03/2019] [Accepted: 04/04/2019] [Indexed: 12/16/2022]
Abstract
Limited regenerative capacity of the nervous system makes treating traumatic nerve injuries with conventional polymer-based nerve grafting a challenging task. Consequently, utilizing natural polymers and biomimetic topologies became obvious strategies for nerve conduit designs. As a bioinspired natural polymer from a marine organism, mussel adhesive proteins (MAPs) fused with biofunctional peptides from extracellular matrix (ECM) were engineered for accelerated nerve regeneration by enhancing cell adhesion, proliferation, neural differentiation, and neurite formation. To physically promote contact guidance of neural and Schwann cells and to achieve guided nerve regeneration, MAP was fabricated into an electrospun aligned nanofiber conduit by introducing synthetic polymer poly(lactic-co-glycolic acid) (PLGA) to control solubility and mechanical property. In vitro and in vivo experiments demonstrated that the multi-dimensional tactics of combining adhesiveness from MAP, integrin-mediated interaction from ECM peptides (in particular, IKVAV derived from laminin α1 chain), and contact guidance from aligned nanofibers synergistically accelerated functional nerve regeneration. Thus, MAP-based multi-dimensional approach provides new opportunities for neural regenerative applications including nerve grafting. STATEMENT OF SIGNIFICANCE: Findings in neural regeneration indicate that a bioinspired polymer-based nerve conduit design should harmoniously constitute various factors, such as biocompatibility, neurotrophic molecule, biodegradability, and contact guidance. Here, we engineered three fusion proteins of mussel-derived adhesive protein with ECM-derived biofunctional peptides to simultaneously provide biocompatibility and integrin-based interactions. In addition, a fabrication of robust aligned nanofiber conduits containing the fusion proteins realized suitable biodegradability and contact guidance. Thus, our multi-dimensional strategy on conduit design provided outstanding biocompatibility, biodegradability, integrin-interaction, and contact guidance to achieve an accelerated functional nerve regeneration. We believe that our bioengineered mussel adhesive protein-based multi-dimensional strategy would offer new insights into the design of nerve tissue engineering biomaterials.
Collapse
|
47
|
Becker K, Kegler K, von Altrock A, Kuchelmeister K, Baumgärtner W, Wohlsein P. Cutaneous Pigmented Neurofibroma in a Pig - Morphology and Immunohistochemical Profile. J Comp Pathol 2019; 168:25-29. [PMID: 31103055 DOI: 10.1016/j.jcpa.2019.03.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2018] [Revised: 03/01/2019] [Accepted: 03/11/2019] [Indexed: 01/28/2023]
Abstract
Peripheral nerve sheath tumours are rare in pigs. In the present case, a juvenile female hybrid pig showed a solitary, pigmented, cutaneous mass. Histologically, it consisted of clustered melanin-laden, epithelioid cells as well as spindle cells forming bundles and nodules. The latter were surrounded by perineurial-like cells. Single Wagner-Meissner-like corpuscles were present. Immunohistochemically, the epithelioid cells expressed S100 protein, melan A and the p75 neurotrophin receptor (p75NTR). The spindle cells expressed S100, sex determining region Y-box 2, p75NTR, Krox20, growth associated protein 43 and glial fibrillary acidic protein. Perineurial-like cells were positive for p75NTR, α-smooth muscle actin and cytokeratin. Taken together, the histological und immunohistochemical findings support the diagnosis of a cutaneous pigmented neurofibroma.
Collapse
Affiliation(s)
- K Becker
- Department of Pathology, Germany
| | - K Kegler
- Department of Pathology, Germany
| | - A von Altrock
- Clinic for Swine, Small Ruminants, Forensic Medicine and Ambulatory Service, University of Veterinary Medicine, Hannover, Germany
| | - K Kuchelmeister
- Department of Neuropathology, University Hospital, Medical School, Bonn, Germany
| | | | | |
Collapse
|
48
|
Bowles KR, Tcw J, Qian L, Jadow BM, Goate AM. Reduced variability of neural progenitor cells and improved purity of neuronal cultures using magnetic activated cell sorting. PLoS One 2019; 14:e0213374. [PMID: 30917153 PMCID: PMC6436701 DOI: 10.1371/journal.pone.0213374] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2018] [Accepted: 02/19/2019] [Indexed: 02/07/2023] Open
Abstract
Genetic and epigenetic variability between iPSC-derived neural progenitor cells (NPCs) combined with differences in investigator technique and selection protocols contributes to variability between NPC lines, which subsequently impacts the quality of differentiated neuronal cultures. We therefore sought to develop an efficient method to reduce this variability in order to improve the purity of NPC and neuronal cultures. Here, we describe a magnetic activated cell sorting (MACS) method for enriching NPC cultures for CD271-/CD133+ cells at both early (<2–3) and late (>10) passage. MACS results in a similar sorting efficiency to fluorescence activated cell sorting (FACS), while achieving an increased yield of live cells and reduced cellular stress. Furthermore, neurons derived from MACS NPCs showed greater homogeneity between cell lines compared to those derived from unsorted NPCs. We conclude that MACS is a cheap technique for incorporation into standard NPC differentiation and maintenance protocols in order to improve culture homogeneity and consistency.
Collapse
Affiliation(s)
- Kathryn R Bowles
- Department of Neuroscience & Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States of America.,Ronald M. Loeb Center for Alzheimer's disease, Icahn School of Medicine at Mount Sinai, New York, NY, United States of America
| | - Julia Tcw
- Department of Neuroscience & Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States of America.,Ronald M. Loeb Center for Alzheimer's disease, Icahn School of Medicine at Mount Sinai, New York, NY, United States of America
| | - Lu Qian
- Department of Neuroscience & Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States of America.,Ronald M. Loeb Center for Alzheimer's disease, Icahn School of Medicine at Mount Sinai, New York, NY, United States of America
| | - Benjamin M Jadow
- Department of Neuroscience & Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States of America.,Ronald M. Loeb Center for Alzheimer's disease, Icahn School of Medicine at Mount Sinai, New York, NY, United States of America
| | - Alison M Goate
- Department of Neuroscience & Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States of America.,Ronald M. Loeb Center for Alzheimer's disease, Icahn School of Medicine at Mount Sinai, New York, NY, United States of America.,Department of Genetics and Genomic Sciences, Icahn Institute of Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY, United States of America
| |
Collapse
|
49
|
Jessen KR, Mirsky R. Schwann Cell Precursors; Multipotent Glial Cells in Embryonic Nerves. Front Mol Neurosci 2019; 12:69. [PMID: 30971890 PMCID: PMC6443887 DOI: 10.3389/fnmol.2019.00069] [Citation(s) in RCA: 105] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2018] [Accepted: 03/04/2019] [Indexed: 12/20/2022] Open
Abstract
The cells of the neural crest, often referred to as neural crest stem cells, give rise to a number of sub-lineages, one of which is Schwann cells, the glial cells of peripheral nerves. Crest cells transform to adult Schwann cells through the generation of two well defined intermediate stages, the Schwann cell precursors (SCP) in early embryonic nerves, and immature Schwann cells (iSch) in late embryonic and perinatal nerves. SCP are formed when neural crest cells enter nascent nerves and form intimate relationships with axons, a diagnostic feature of glial cells. This involves large-scale changes in gene expression, including the activation of established glial cell markers. Like early glia in the CNS, radial glia, SCP retain developmental multipotency and contribute to other crest-derived lineages during embryonic development. SCP, as well as closely related cells termed boundary cap cells, and later stages of the Schwann cell lineage have all been implicated as the tumor initiating cell in NF1 associated neurofibromas. iSch are formed from SCP in a process that involves the appearance of additional differentiation markers, autocrine survival circuits, cellular elongation, a formation of endoneurial connective tissue and basal lamina. Finally, in peri- and post-natal nerves, iSch are reversibly induced by axon-associated signals to form the myelin and non-myelin Schwann cells of adult nerves. This review article discusses early Schwann cell development in detail and describes a large number of molecular signaling systems that control glial development in embryonic nerves.
Collapse
Affiliation(s)
- Kristjan R. Jessen
- Department of Cell and Developmental Biology, University College London, London, United Kingdom
| | | |
Collapse
|
50
|
Kamil K, Yazid MD, Idrus RBH, Das S, Kumar J. Peripheral Demyelinating Diseases: From Biology to Translational Medicine. Front Neurol 2019; 10:87. [PMID: 30941082 PMCID: PMC6433847 DOI: 10.3389/fneur.2019.00087] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Accepted: 01/22/2019] [Indexed: 12/30/2022] Open
Abstract
Demyelinating diseases represent a spectrum of disorders that impose significant burden on global economy and society. Generally, the prognosis of these diseases is poor and there is no available cure. In recent decades, research has shed some light on the biology and physiology of Schwann cells and its neuroprotective effects in the peripheral nervous system (PNS). Insults to the PNS by various infectious agents, genetic predisposition and immune-related mechanisms jeopardize Schwann cell functions and cause demyelination. To date, there are no effective and reliable biomarkers for PNS-related diseases. Here, we aim to review the following: pathogenesis of various types of peripheral demyelinating diseases such as Guillain-Barre syndrome, Chronic Inflammatory Demyelinating Polyradiculoneuropathy, Anti-Myelin Associated Glycoprotein Neuropathy, POEMS syndrome, and Charcot-Marie-Tooth disease; emerging novel biomarkers for peripheral demyelinating diseases, and Schwann cell associated markers for demyelination.
Collapse
Affiliation(s)
- Khidhir Kamil
- Department of Physiology, Faculty of Medicine, Universiti Kebangsaan Malaysia Medical Centre, Kuala Lumpur, Malaysia
| | - Muhammad Dain Yazid
- Tissue Engineering Centre, Faculty of Medicine, Universiti Kebangsaan Malaysia Medical Centre, Kuala Lumpur, Malaysia
| | - Ruszymah Bt Hj Idrus
- Department of Physiology, Faculty of Medicine, Universiti Kebangsaan Malaysia Medical Centre, Kuala Lumpur, Malaysia
| | - Srijit Das
- Department of Anatomy, Faculty of Medicine, Universiti Kebangsaan Malaysia Medical Centre, Kuala Lumpur, Malaysia
| | - Jaya Kumar
- Department of Physiology, Faculty of Medicine, Universiti Kebangsaan Malaysia Medical Centre, Kuala Lumpur, Malaysia
| |
Collapse
|