1
|
Dhar A, Moinuddin FM, Zamanian CA, Sharar AD, Dominari A, Graepel S, Windebank AJ, Bydon M. SOX Genes in Spinal Cord Injury: Redefining Neural Stem Cell Regeneration Strategies. Mol Neurobiol 2025:10.1007/s12035-025-04882-w. [PMID: 40156684 DOI: 10.1007/s12035-025-04882-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Accepted: 03/21/2025] [Indexed: 04/01/2025]
Abstract
The study design is literature review. The sex-determining region Y gene (SRY)-related high mobility group box (HMG)-box (SOX) gene family has primarily been associated with neural development and sex determination and is a key component of human embryonic development. Recent studies on zebrafish models have demonstrated that the unique ability of the latter for central nervous tissue (CNS) repair following injury is largely mediated by SOX genes. Given that efforts aimed at the structural regeneration and functional restoration of neural tissue still represent a major therapeutic challenge in patients suffering CNS injury, these findings have initiated a discussion regarding the development of novel therapeutic strategies for SCI focusing on neural tissue regeneration. Spinal cord injury (SCI), in particular, represents a field that could greatly benefit from studies related to the function of the SOX genes. Neuro-informatics Laboratory, Mayo Clinic, Rochester, MN. A literature review was conducted, with a focus on SOX gene that has been described in the experimental studies of SCI. In this review, the existing evidence linking the SOX gene family to the pathophysiology of SCI is summarized, and future research steps regarding the potential implications of the SOX genes in neurological recovery following SCI are discussed, especially focusing on highlighting potential therapeutic targets. The potential implications of the latter could play a crucial role in future efforts to advance the treatment approaches to SCI.
Collapse
Affiliation(s)
- Ashis Dhar
- Mayo Clinic Neuro-Informatics Laboratory, Department of Neurosurgery, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
- Department of Neurological Surgery, Mayo Clinic, Rochester, MN, USA
| | - F M Moinuddin
- Mayo Clinic Neuro-Informatics Laboratory, Department of Neurosurgery, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
- Department of Neurological Surgery, Mayo Clinic, Rochester, MN, USA
| | - Cameron A Zamanian
- Mayo Clinic Neuro-Informatics Laboratory, Department of Neurosurgery, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
- Department of Neurological Surgery, Mayo Clinic, Rochester, MN, USA
| | - Ahnaf Dil Sharar
- Mayo Clinic Neuro-Informatics Laboratory, Department of Neurosurgery, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
- Department of Neurological Surgery, Mayo Clinic, Rochester, MN, USA
| | - Asimina Dominari
- Mayo Clinic Neuro-Informatics Laboratory, Department of Neurosurgery, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
- Department of Neurological Surgery, Mayo Clinic, Rochester, MN, USA
| | - Stephen Graepel
- Department of Neurological Surgery, Mayo Clinic, Rochester, MN, USA
| | | | - Mohamad Bydon
- Mayo Clinic Neuro-Informatics Laboratory, Department of Neurosurgery, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA.
- Department of Neurological Surgery, Mayo Clinic, Rochester, MN, USA.
| |
Collapse
|
2
|
Kandror EK, Wang A, Carriere M, Peterson A, Liao W, Tjärnberg A, Fung JH, Mahbubani KT, Loper J, Pangburn W, Xu Y, Saeb-Parsy K, Rabadan R, Maniatis T, Rizvi AH. Enhancer Dynamics and Spatial Organization Drive Anatomically Restricted Cellular States in the Human Spinal Cord. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.10.632483. [PMID: 39829819 PMCID: PMC11741326 DOI: 10.1101/2025.01.10.632483] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 01/22/2025]
Abstract
Here, we report the spatial organization of RNA transcription and associated enhancer dynamics in the human spinal cord at single-cell and single-molecule resolution. We expand traditional multiomic measurements to reveal epigenetically poised and bivalent active transcriptional enhancer states that define cell type specification. Simultaneous detection of chromatin accessibility and histone modifications in spinal cord nuclei reveals previously unobserved cell-type specific cryptic enhancer activity, in which transcriptional activation is uncoupled from chromatin accessibility. Such cryptic enhancers define both stable cell type identity and transitions between cells undergoing differentiation. We also define glial cell gene regulatory networks that reorganize along the rostrocaudal axis, revealing anatomical differences in gene regulation. Finally, we identify the spatial organization of cells into distinct cellular organizations and address the functional significance of this observation in the context of paracrine signaling. We conclude that cellular diversity is best captured through the lens of enhancer state and intercellular interactions that drive transitions in cellular state. This study provides fundamental insights into the cellular organization of the healthy human spinal cord.
Collapse
Affiliation(s)
- Elena K. Kandror
- Department of Neuroscience and Waisman Center, University of Wisconsin-Madison
| | - Anqi Wang
- Program for Mathematical Genomics, Department of Systems Biology, Columbia University Medical Center
| | | | - Alexis Peterson
- Department of Neuroscience and Waisman Center, University of Wisconsin-Madison
| | | | - Andreas Tjärnberg
- Department of Neuroscience and Waisman Center, University of Wisconsin-Madison
| | - Jun Hou Fung
- Program for Mathematical Genomics, Department of Systems Biology, Columbia University Medical Center
| | - Krishnaa T. Mahbubani
- Cambridge Biorepository for Translational Medicine, Cambridge NIHR Biomedical Research Centre, Cambridge, UK
- Department of Surgery, University of Cambridge, Cambridge, UK
- Department of Haematology, Cambridge Stem Cell Institute, Cambridge, UK
| | - Jackson Loper
- Department of Statistics, University of Michigan Ann Arbor
| | - William Pangburn
- Zuckerman Mind Brain Behavior Institute and Department of Biochemistry and Molecular Biophysics, Columbia University Medical Center
| | - Yuchen Xu
- Department of Neuroscience and Waisman Center, University of Wisconsin-Madison
| | - Kourosh Saeb-Parsy
- Cambridge Biorepository for Translational Medicine, Cambridge NIHR Biomedical Research Centre, Cambridge, UK
- Department of Surgery, University of Cambridge, Cambridge, UK
| | - Raul Rabadan
- Program for Mathematical Genomics, Department of Systems Biology, Columbia University Medical Center
| | - Tom Maniatis
- New York Genome Center
- Zuckerman Mind Brain Behavior Institute and Department of Biochemistry and Molecular Biophysics, Columbia University Medical Center
| | - Abbas H. Rizvi
- Department of Neuroscience and Waisman Center, University of Wisconsin-Madison
- Lead contact
| |
Collapse
|
3
|
Luciani M, Garsia C, Beretta S, Cifola I, Peano C, Merelli I, Petiti L, Miccio A, Meneghini V, Gritti A. Human iPSC-derived neural stem cells displaying radial glia signature exhibit long-term safety in mice. Nat Commun 2024; 15:9433. [PMID: 39487141 PMCID: PMC11530573 DOI: 10.1038/s41467-024-53613-7] [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: 08/31/2023] [Accepted: 10/17/2024] [Indexed: 11/04/2024] Open
Abstract
Human induced pluripotent stem cell-derived neural stem/progenitor cells (hiPSC-NSCs) hold promise for treating neurodegenerative and demyelinating disorders. However, comprehensive studies on their identity and safety remain limited. In this study, we demonstrate that hiPSC-NSCs adopt a radial glia-associated signature, sharing key epigenetic and transcriptional characteristics with human fetal neural stem cells (hfNSCs) while exhibiting divergent profiles from glioblastoma stem cells. Long-term transplantation studies in mice showed robust and stable engraftment of hiPSC-NSCs, with predominant differentiation into glial cells and no evidence of tumor formation. Additionally, we identified the Sterol Regulatory Element Binding Transcription Factor 1 (SREBF1) as a regulator of astroglial differentiation in hiPSC-NSCs. These findings provide valuable transcriptional and epigenetic reference datasets to prospectively define the maturation stage of NSCs derived from different hiPSC sources and demonstrate the long-term safety of hiPSC-NSCs, reinforcing their potential as a viable alternative to hfNSCs for clinical applications.
Collapse
Affiliation(s)
- Marco Luciani
- San Raffaele Telethon Institute for Gene Therapy, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Scientific Institute, Milan, Italy
| | - Chiara Garsia
- San Raffaele Telethon Institute for Gene Therapy, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Scientific Institute, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| | - Stefano Beretta
- San Raffaele Telethon Institute for Gene Therapy, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Scientific Institute, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| | - Ingrid Cifola
- Institute for Biomedical Technologies (ITB), National Research Council (CNR), via F.lli Cervi 93, 20054 Segrate, Milan, Italy
| | - Clelia Peano
- Institute of Genetics and Biomedical Research, UoS of Milan, National Research Council, Rozzano, Milan, Italy
- Human Technopole, Via Rita Levi Montalcini 1, Milan, Italy
| | - Ivan Merelli
- San Raffaele Telethon Institute for Gene Therapy, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Scientific Institute, Milan, Italy
| | - Luca Petiti
- Institute for Biomedical Technologies (ITB), National Research Council (CNR), via F.lli Cervi 93, 20054 Segrate, Milan, Italy
| | - Annarita Miccio
- IMAGINE Institute, Université de Paris, Sorbonne Paris Cité, Paris, France
| | - Vasco Meneghini
- San Raffaele Telethon Institute for Gene Therapy, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Scientific Institute, Milan, Italy.
- Vita-Salute San Raffaele University, Milan, Italy.
| | - Angela Gritti
- San Raffaele Telethon Institute for Gene Therapy, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Scientific Institute, Milan, Italy.
- Vita-Salute San Raffaele University, Milan, Italy.
| |
Collapse
|
4
|
England SJ, Campbell PC, Banerjee S, Bates RL, Grieb G, Fancher WF, Lewis KE. Transcriptional regulators with broad expression in the zebrafish spinal cord. Dev Dyn 2024; 253:1036-1055. [PMID: 38850245 DOI: 10.1002/dvdy.717] [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: 01/23/2024] [Revised: 04/12/2024] [Accepted: 05/15/2024] [Indexed: 06/10/2024] Open
Abstract
BACKGROUND The spinal cord is a crucial part of the vertebrate CNS, controlling movements and receiving and processing sensory information from the trunk and limbs. However, there is much we do not know about how this essential organ develops. Here, we describe expression of 21 transcription factors and one transcriptional regulator in zebrafish spinal cord. RESULTS We analyzed the expression of aurkb, foxb1a, foxb1b, her8a, homeza, ivns1abpb, mybl2b, myt1a, nr2f1b, onecut1, sall1a, sall3a, sall3b, sall4, sox2, sox19b, sp8b, tsc22d1, wdhd1, zfhx3b, znf804a, and znf1032 in wild-type and MIB E3 ubiquitin protein ligase 1 zebrafish embryos. While all of these genes are broadly expressed in spinal cord, they have distinct expression patterns from one another. Some are predominantly expressed in progenitor domains, and others in subsets of post-mitotic cells. Given the conservation of spinal cord development, and the transcription factors and transcriptional regulators that orchestrate it, we expect that these genes will have similar spinal cord expression patterns in other vertebrates, including mammals and humans. CONCLUSIONS Our data identify 22 different transcriptional regulators that are strong candidates for playing different roles in spinal cord development. For several of these genes, this is the first published description of their spinal cord expression.
Collapse
Affiliation(s)
| | - Paul C Campbell
- Department of Biology, Syracuse University, Syracuse, New York, USA
| | - Santanu Banerjee
- Biological Sciences Department, SUNY-Cortland, Cortland, New York, USA
| | - Richard L Bates
- Department of Biology, Syracuse University, Syracuse, New York, USA
| | - Ginny Grieb
- Department of Biology, Syracuse University, Syracuse, New York, USA
| | | | | |
Collapse
|
5
|
Poon F, Sambathkumar R, Korytnikov R, Aghazadeh Y, Oakie A, Misra PS, Sarangi F, Nostro MC. Tankyrase inhibition promotes endocrine commitment of hPSC-derived pancreatic progenitors. Nat Commun 2024; 15:8754. [PMID: 39384787 PMCID: PMC11464881 DOI: 10.1038/s41467-024-53068-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Accepted: 09/27/2024] [Indexed: 10/11/2024] Open
Abstract
Human pluripotent stem cells (hPSCs) have the potential to differentiate into various cell types, including pancreatic insulin-producing β cells, which are crucial for developing therapies for diabetes. However, current methods for directing hPSC differentiation towards pancreatic β-like cells are often inefficient and produce cells that do not fully resemble the native counterparts. Here, we report that highly selective tankyrase inhibitors, such as WIKI4, significantly enhances pancreatic differentiation from hPSCs. Our results show that WIKI4 promotes the formation of pancreatic progenitors that give rise to islet-like cells with improved β-like cell frequencies and glucose responsiveness compared to our standard cultures. These findings not only advance our understanding of pancreatic development, but also provide a promising new tool for generating pancreatic cells for research and potential therapeutic applications.
Collapse
Affiliation(s)
- Frankie Poon
- McEwen Stem Cell Institute, University Health Network, Toronto, ON, M5G 1L7, Canada
- Department of Physiology, University of Toronto, Toronto, ON, M5S 1A8, Canada
- Sana Biotechnology, 300 Technology Square, Cambridge, MA, 02139, USA
| | - Rangarajan Sambathkumar
- McEwen Stem Cell Institute, University Health Network, Toronto, ON, M5G 1L7, Canada
- Allarta Life Science Inc., 1280 Main Street West, Hamilton, ON, L8S 4L8, Canada
| | - Roman Korytnikov
- McEwen Stem Cell Institute, University Health Network, Toronto, ON, M5G 1L7, Canada
- Department of Physiology, University of Toronto, Toronto, ON, M5S 1A8, Canada
| | - Yasaman Aghazadeh
- McEwen Stem Cell Institute, University Health Network, Toronto, ON, M5G 1L7, Canada
- Montreal Clinical Research Institute (IRCM), University of Montreal, Department of Medicine, Montreal, H2W 1R7, QC, Canada
| | - Amanda Oakie
- McEwen Stem Cell Institute, University Health Network, Toronto, ON, M5G 1L7, Canada
| | - Paraish S Misra
- McEwen Stem Cell Institute, University Health Network, Toronto, ON, M5G 1L7, Canada
- Department of Physiology, University of Toronto, Toronto, ON, M5S 1A8, Canada
- Department of Medicine, University of Toronto, Toronto, ON, M5S 1A8, Canada
| | - Farida Sarangi
- McEwen Stem Cell Institute, University Health Network, Toronto, ON, M5G 1L7, Canada
| | - M Cristina Nostro
- McEwen Stem Cell Institute, University Health Network, Toronto, ON, M5G 1L7, Canada.
- Department of Physiology, University of Toronto, Toronto, ON, M5S 1A8, Canada.
| |
Collapse
|
6
|
Galichet C, Rizzoti K, Lovell-Badge R. Hypopituitarism in Sox3 null mutants correlates with altered NG2-glia in the median eminence and is influenced by aspirin and gut microbiota. PLoS Genet 2024; 20:e1011395. [PMID: 39325695 PMCID: PMC11426531 DOI: 10.1371/journal.pgen.1011395] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Accepted: 08/13/2024] [Indexed: 09/28/2024] Open
Abstract
The median eminence (ME), located at the base of the hypothalamus, is an essential centre of information exchange between the brain and the pituitary. We and others previously showed that mutations and duplications affecting the transcription factor SOX3/Sox3 result in hypopituitarism, and this is likely of hypothalamic origin. We demonstrate here that the absence of Sox3 predominantly affects the ME with phenotypes that first occur in juvenile animals, despite the embryonic onset of SOX3 expression. In the pituitary, reduction in hormone levels correlates with a lack of endocrine cell maturation. In parallel, ME NG2-glia renewal and oligodendrocytic differentiation potential are affected. We further show that low-dose aspirin treatment, which is known to affect NG2-glia, or changes in gut microbiota, rescue both proliferative defects and hypopituitarism in Sox3 mutants. Our study highlights a central role of NG2-glia for ME function during a transitional period of post-natal development and indicates their sensitivity to extrinsic signals.
Collapse
Affiliation(s)
- Christophe Galichet
- Stem Cell Biology and Developmental Genetics Lab, The Francis Crick Institute, London, United Kingdom
- Neurobiological Research Facility, UCL Sainsbury Wellcome Centre for Neural Circuits and Behaviour, London, United Kingdom
| | - Karine Rizzoti
- Stem Cell Biology and Developmental Genetics Lab, The Francis Crick Institute, London, United Kingdom
| | - Robin Lovell-Badge
- Stem Cell Biology and Developmental Genetics Lab, The Francis Crick Institute, London, United Kingdom
| |
Collapse
|
7
|
Damuth DL, Cunningham DD, Silva EM. Sox21 homeologs autoregulate expression levels to control progression through neurogenesis. Genesis 2024; 62:e23612. [PMID: 39054872 PMCID: PMC11584151 DOI: 10.1002/dvg.23612] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Revised: 05/29/2024] [Accepted: 06/10/2024] [Indexed: 07/27/2024]
Abstract
The SRY HMG box transcription factor Sox21 plays multiple critical roles in neurogenesis, with its function dependent on concentration and developmental stage. In the allotetraploid Xenopus laevis, there are two homeologs of sox21, namely sox21.S and sox21.L. Previous studies focused on Sox21.S, but its amino acid sequence is divergent, lacking conserved poly-A stretches and bearing more similarity with ancestral homologs. In contrast, Sox21.L shares higher sequence similarity with mouse and chick Sox21. To determine if Sox21.S and Sox21.L have distinct functions, we conducted gain and loss-of-function studies in Xenopus embryos. Our studies revealed that Sox21.S and Sox21.L are functionally redundant, but Sox21.L is more effective at driving changes than Sox21.S. These results also support our earlier findings in ectodermal explants, demonstrating that Sox21 function is dose-dependent. While Sox21 is necessary for primary neuron formation, high levels prevent their formation. Strikingly, these proteins autoregulate, with high levels of Sox21.L reducing sox21.S and sox21.L mRNA levels, and decreased Sox21.S promoting increased expression of sox21.L. Our findings shed light on the intricate concentration-dependent roles of Sox21 homeologs in Xenopus neurogenesis.
Collapse
Affiliation(s)
- Dillon L Damuth
- Department of Biology, Georgetown University, Washington, DC, USA
| | | | - Elena M Silva
- Department of Biology, Georgetown University, Washington, DC, USA
| |
Collapse
|
8
|
Kornfeld SF, Cummings SE, Yaworski R, De Repentigny Y, Gagnon S, Zandee S, Fathi S, Prat A, Kothary R. Loss of miR-145 promotes remyelination and functional recovery in a model of chronic central demyelination. Commun Biol 2024; 7:813. [PMID: 38965401 PMCID: PMC11224363 DOI: 10.1038/s42003-024-06513-x] [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: 09/23/2020] [Accepted: 06/27/2024] [Indexed: 07/06/2024] Open
Abstract
Strategies for treating progressive multiple sclerosis (MS) remain limited. Here, we found that miR-145-5p is overabundant uniquely in chronic lesion tissues from secondary progressive MS patients. We induced both acute and chronic demyelination in miR-145 knockout mice to determine its contributions to remyelination failure. Following acute demyelination, no advantage to miR-145 loss could be detected. However, after chronic demyelination, animals with miR-145 loss demonstrated increased remyelination and functional recovery, coincident with altered presence of astrocytes and microglia within the corpus callosum relative to wild-type animals. This improved response in miR-145 knockout animals coincided with a pathological upregulation of miR-145-5p in wild-type animals with chronic cuprizone exposure, paralleling human chronic lesions. Furthermore, miR-145 overexpression specifically in oligodendrocytes (OLs) severely stunted differentiation and negatively impacted survival. RNAseq analysis showed altered transcriptome in these cells with downregulated major pathways involved in myelination. Our data suggest that pathological accumulation of miR-145-5p is a distinctive feature of chronic demyelination and is strongly implicated in the failure of remyelination, possibly due to the inhibition of OL differentiation together with alterations in other glial cells. This is mirrored in chronic MS lesions, and thus miR-145-5p serves as a potential relevant therapeutic target in progressive forms of MS.
Collapse
Affiliation(s)
- Samantha F Kornfeld
- Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, ON, K1H 8L6, Canada
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON, K1H 8M5, Canada
| | - Sarah E Cummings
- Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, ON, K1H 8L6, Canada
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON, K1H 8M5, Canada
| | - Rebecca Yaworski
- Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, ON, K1H 8L6, Canada
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON, K1H 8M5, Canada
| | - Yves De Repentigny
- Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, ON, K1H 8L6, Canada
| | - Sabrina Gagnon
- Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, ON, K1H 8L6, Canada
| | - Stephanie Zandee
- Neuroimmunology Unit and Multiple Sclerosis Clinic, The Research Center of the Centre Hospitalier de l'Université de Montréal (CRCHUM), Department of Neuroscience, Faculty of Medicine, Université de Montréal, Montréal, QC, Canada
| | - Samaneh Fathi
- Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, ON, K1H 8L6, Canada
| | - Alexandre Prat
- Neuroimmunology Unit and Multiple Sclerosis Clinic, The Research Center of the Centre Hospitalier de l'Université de Montréal (CRCHUM), Department of Neuroscience, Faculty of Medicine, Université de Montréal, Montréal, QC, Canada
| | - Rashmi Kothary
- Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, ON, K1H 8L6, Canada.
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON, K1H 8M5, Canada.
- Department of Medicine, University of Ottawa, Ottawa, ON, K1H 8M5, Canada.
- Centre for Neuromuscular Disease, University of Ottawa, Ottawa, ON, K1H 8M5, Canada.
| |
Collapse
|
9
|
Rupprecht J, Reiprich S, Baroti T, Christoph C, Sock E, Fröb F, Wegner M. Transcription Factors Sox2 and Sox3 Directly Regulate the Expression of Genes Involved in the Onset of Oligodendrocyte Differentiation. Cells 2024; 13:935. [PMID: 38891067 PMCID: PMC11172379 DOI: 10.3390/cells13110935] [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: 04/29/2024] [Revised: 05/17/2024] [Accepted: 05/28/2024] [Indexed: 06/21/2024] Open
Abstract
Rapid information processing in the central nervous system requires the myelination of axons by oligodendrocytes. The transcription factor Sox2 and its close relative Sox3 redundantly regulate the development of myelin-forming oligodendrocytes, but little is known about the underlying molecular mechanisms. Here, we characterized the expression profile of cultured oligodendroglial cells during early differentiation and identified Bcas1, Enpp6, Zfp488 and Nkx2.2 as major downregulated genes upon Sox2 and Sox3 deletion. An analysis of mice with oligodendrocyte-specific deletion of Sox2 and Sox3 validated all four genes as downstream targets in vivo. Additional functional assays identified regulatory regions in the vicinity of each gene that are responsive to and bind both Sox proteins. Bcas1, Enpp6, Zfp488 and Nkx2.2 therefore likely represent direct target genes and major effectors of Sox2 and Sox3. Considering the preferential expression and role of these genes in premyelinating oligodendrocytes, our findings suggest that Sox2 and Sox3 impact oligodendroglial development at the premyelinating stage with Bcas1, Enpp6, Zfp488 and Nkx2.2 as their major effectors.
Collapse
Affiliation(s)
| | | | | | | | | | - Franziska Fröb
- Institut für Biochemie, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Michael Wegner
- Institut für Biochemie, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054 Erlangen, Germany
| |
Collapse
|
10
|
Fang Y, Barrows D, Dabas Y, Carroll T, Singer S, Tap W, Nacev B. ATRX guards against aberrant differentiation in mesenchymal progenitor cells. Nucleic Acids Res 2024; 52:4950-4968. [PMID: 38477352 PMCID: PMC11109985 DOI: 10.1093/nar/gkae160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Revised: 01/19/2024] [Accepted: 03/05/2024] [Indexed: 03/14/2024] Open
Abstract
Alterations in the tumor suppressor ATRX are recurrently observed in mesenchymal neoplasms. ATRX has multiple epigenetic functions including heterochromatin formation and maintenance and regulation of transcription through modulation of chromatin accessibility. Here, we show in murine mesenchymal progenitor cells (MPCs) that Atrx deficiency aberrantly activated mesenchymal differentiation programs. This includes adipogenic pathways where ATRX loss induced expression of adipogenic transcription factors and enhanced adipogenic differentiation in response to differentiation stimuli. These changes are linked to loss of heterochromatin near mesenchymal lineage genes together with increased chromatin accessibility and gains of active chromatin marks. We additionally observed depletion of H3K9me3 at transposable elements, which are derepressed including near mesenchymal genes where they could serve as regulatory elements. Finally, we demonstrated that loss of ATRX in a mesenchymal malignancy, undifferentiated pleomorphic sarcoma, results in similar epigenetic disruption and de-repression of transposable elements. Together, our results reveal a role for ATRX in maintaining epigenetic states and transcriptional repression in mesenchymal progenitors and tumor cells and in preventing aberrant differentiation in the progenitor context.
Collapse
Affiliation(s)
- Yan Fang
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY10065, USA
- Laboratory of Chromatin Biology and Epigenetics, The Rockefeller University, New York, NY 10065, USA
| | - Douglas Barrows
- Bioinformatics Resource Center, The Rockefeller University, New York, NY10065, USA
| | - Yakshi Dabas
- Laboratory of Chromatin Biology and Epigenetics, The Rockefeller University, New York, NY 10065, USA
| | - Thomas S Carroll
- Bioinformatics Resource Center, The Rockefeller University, New York, NY10065, USA
| | - Sam Singer
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY10065, USA
| | - William D Tap
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY10065, USA
| | - Benjamin A Nacev
- Department of Medicine, University of Pittsburgh, Pittsburgh, PA 15213, USA
- Department of Pathology, University of Pittsburgh, Pittsburgh, PA 15213, USA
- UPMC Hillman Cancer Center, Pittsburgh, PA 15213, USA
| |
Collapse
|
11
|
England SJ, Campbell PC, Banerjee S, Bates RL, Grieb G, Fancher WF, Lewis KE. Transcriptional Regulators with Broad Expression in the Zebrafish Spinal Cord. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.14.580357. [PMID: 38405913 PMCID: PMC10888778 DOI: 10.1101/2024.02.14.580357] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/27/2024]
Abstract
Background The spinal cord is a crucial part of the vertebrate CNS, controlling movements and receiving and processing sensory information from the trunk and limbs. However, there is much we do not know about how this essential organ develops. Here, we describe expression of 21 transcription factors and one transcriptional regulator in zebrafish spinal cord. Results We analyzed the expression of aurkb, foxb1a, foxb1b, her8a, homeza, ivns1abpb, mybl2b, myt1a, nr2f1b, onecut1, sall1a, sall3a, sall3b, sall4, sox2, sox19b, sp8b, tsc22d1, wdhd1, zfhx3b, znf804a, and znf1032 in wild-type and MIB E3 ubiquitin protein ligase 1 zebrafish embryos. While all of these genes are broadly expressed in spinal cord, they have distinct expression patterns from one another. Some are predominantly expressed in progenitor domains, and others in subsets of post-mitotic cells. Given the conservation of spinal cord development, and the transcription factors and transcriptional regulators that orchestrate it, we expect that these genes will have similar spinal cord expression patterns in other vertebrates, including mammals and humans. Conclusions Our data identify 22 different transcriptional regulators that are strong candidates for playing different roles in spinal cord development. For several of these genes, this is the first published description of their spinal cord expression.
Collapse
Affiliation(s)
- Samantha J. England
- Department of Biology, Syracuse University, 107 College Place, Syracuse, NY 13244, USA
| | - Paul C. Campbell
- Department of Biology, Syracuse University, 107 College Place, Syracuse, NY 13244, USA
| | - Santanu Banerjee
- Biological Sciences Department, SUNY-Cortland, Cortland, NY 13045, USA
| | - Richard L. Bates
- Department of Biology, Syracuse University, 107 College Place, Syracuse, NY 13244, USA
| | - Ginny Grieb
- Department of Biology, Syracuse University, 107 College Place, Syracuse, NY 13244, USA
| | - William F. Fancher
- Department of Biology, Syracuse University, 107 College Place, Syracuse, NY 13244, USA
| | - Katharine E. Lewis
- Department of Biology, Syracuse University, 107 College Place, Syracuse, NY 13244, USA
| |
Collapse
|
12
|
Baričević Z, Pongrac M, Ivaničić M, Hreščak H, Tomljanović I, Petrović A, Cojoc D, Mladinic M, Ban J. SOX2 and SOX9 Expression in Developing Postnatal Opossum ( Monodelphis domestica) Cortex. Biomolecules 2024; 14:70. [PMID: 38254670 PMCID: PMC10813269 DOI: 10.3390/biom14010070] [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: 11/24/2023] [Revised: 12/30/2023] [Accepted: 12/31/2023] [Indexed: 01/24/2024] Open
Abstract
(1) Background: Central nervous system (CNS) development is characterized by dynamic changes in cell proliferation and differentiation. Key regulators of these transitions are the transcription factors such as SOX2 and SOX9. SOX2 is involved in the maintenance of progenitor cell state and neural stem cell multipotency, while SOX9, expressed in neurogenic niches, plays an important role in neuron/glia switch with predominant expression in astrocytes in the adult brain. (2) Methods: To validate SOX2 and SOX9 expression patterns in developing opossum (Monodelphis domestica) cortex, we used immunohistochemistry (IHC) and the isotropic fractionator method on fixed cortical tissue from comparable postnatal ages, as well as dissociated primary neuronal cultures. (3) Results: Neurons positive for both neuronal (TUJ1 or NeuN) and stem cell (SOX2) markers were identified, and their presence was confirmed with all methods and postnatal age groups (P4-6, P6-18, and P30) analyzed. SOX9 showed exclusive staining in non-neuronal cells, and it was coexpressed with SOX2. (4) Conclusions: The persistence of SOX2 expression in developing cortical neurons of M. domestica during the first postnatal month implies the functional role of SOX2 during neuronal differentiation and maturation, which was not previously reported in opossums.
Collapse
Affiliation(s)
- Zrinko Baričević
- Faculty of Biotechnology and Drug Development, University of Rijeka, Radmile Matejčić 2, 51000 Rijeka, Croatia; (Z.B.); (M.P.); (M.I.); (H.H.); (I.T.); (A.P.); (M.M.)
| | - Marta Pongrac
- Faculty of Biotechnology and Drug Development, University of Rijeka, Radmile Matejčić 2, 51000 Rijeka, Croatia; (Z.B.); (M.P.); (M.I.); (H.H.); (I.T.); (A.P.); (M.M.)
| | - Matea Ivaničić
- Faculty of Biotechnology and Drug Development, University of Rijeka, Radmile Matejčić 2, 51000 Rijeka, Croatia; (Z.B.); (M.P.); (M.I.); (H.H.); (I.T.); (A.P.); (M.M.)
| | - Helena Hreščak
- Faculty of Biotechnology and Drug Development, University of Rijeka, Radmile Matejčić 2, 51000 Rijeka, Croatia; (Z.B.); (M.P.); (M.I.); (H.H.); (I.T.); (A.P.); (M.M.)
| | - Ivana Tomljanović
- Faculty of Biotechnology and Drug Development, University of Rijeka, Radmile Matejčić 2, 51000 Rijeka, Croatia; (Z.B.); (M.P.); (M.I.); (H.H.); (I.T.); (A.P.); (M.M.)
| | - Antonela Petrović
- Faculty of Biotechnology and Drug Development, University of Rijeka, Radmile Matejčić 2, 51000 Rijeka, Croatia; (Z.B.); (M.P.); (M.I.); (H.H.); (I.T.); (A.P.); (M.M.)
| | - Dan Cojoc
- CNR-IOM, Materials Foundry, National Research Council of Italy, 34149 Trieste, Italy;
| | - Miranda Mladinic
- Faculty of Biotechnology and Drug Development, University of Rijeka, Radmile Matejčić 2, 51000 Rijeka, Croatia; (Z.B.); (M.P.); (M.I.); (H.H.); (I.T.); (A.P.); (M.M.)
| | - Jelena Ban
- Faculty of Biotechnology and Drug Development, University of Rijeka, Radmile Matejčić 2, 51000 Rijeka, Croatia; (Z.B.); (M.P.); (M.I.); (H.H.); (I.T.); (A.P.); (M.M.)
| |
Collapse
|
13
|
Wu L, Wang F, Moncman CL, Pandey M, Clarke HA, Frazier HN, Young LE, Gentry MS, Cai W, Thibault O, Sun RC, Andres DA. RIT1 regulation of CNS lipids RIT1 deficiency Alters cerebral lipid metabolism and reduces white matter tract oligodendrocytes and conduction velocities. Heliyon 2023; 9:e20384. [PMID: 37780758 PMCID: PMC10539968 DOI: 10.1016/j.heliyon.2023.e20384] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Revised: 07/21/2023] [Accepted: 09/20/2023] [Indexed: 10/03/2023] Open
Abstract
Oligodendrocytes (OLs) generate lipid-rich myelin membranes that wrap axons to enable efficient transmission of electrical impulses. Using a RIT1 knockout mouse model and in situ high-resolution matrix-assisted laser desorption/ionization mass spectrometry imaging (MALDI-MSI) coupled with MS-based lipidomic analysis to determine the contribution of RIT1 to lipid homeostasis. Here, we report that RIT1 loss is associated with altered lipid levels in the central nervous system (CNS), including myelin-associated lipids within the corpus callosum (CC). Perturbed lipid metabolism was correlated with reduced numbers of OLs, but increased numbers of GFAP+ glia, in the CC, but not in grey matter. This was accompanied by reduced myelin protein expression and axonal conduction deficits. Behavioral analyses revealed significant changes in voluntary locomotor activity and anxiety-like behavior in RIT1KO mice. Together, these data reveal an unexpected role for RIT1 in the regulation of cerebral lipid metabolism, which coincide with altered white matter tract oligodendrocyte levels, reduced axonal conduction velocity, and behavioral abnormalities in the CNS.
Collapse
Affiliation(s)
- Lei Wu
- Department of Molecular and Cellular Biochemistry, College of Medicine, University of Kentucky, KY 40536, USA
| | - Fang Wang
- Department of Molecular and Cellular Biochemistry, College of Medicine, University of Kentucky, KY 40536, USA
| | - Carole L. Moncman
- Department of Molecular and Cellular Biochemistry, College of Medicine, University of Kentucky, KY 40536, USA
| | - Mritunjay Pandey
- Department of Molecular and Cellular Biochemistry, College of Medicine, University of Kentucky, KY 40536, USA
| | - Harrison A. Clarke
- Department of Neuroscience, College of Medicine, University of Kentucky, KY 40536, USA
| | - Hilaree N. Frazier
- Department of Pharmacological and Nutritional Sciences, College of Medicine, University of Kentucky, KY 40536, USA
| | - Lyndsay E.A. Young
- Department of Molecular and Cellular Biochemistry, College of Medicine, University of Kentucky, KY 40536, USA
- Markey Cancer Center, Lexington, KY 40536, USA
| | - Matthew S. Gentry
- Department of Molecular and Cellular Biochemistry, College of Medicine, University of Kentucky, KY 40536, USA
- Markey Cancer Center, Lexington, KY 40536, USA
- Department of Biochemistry and Molecular Biology, University of Florida, College of Medicine, Gainesville, FL 32611, USA
- Center for Advanced Spatial Biomolecule Research, University of Florida, College of Medicine, Gainesville, FL 32611, USA
| | - Weikang Cai
- Department of Biomedical Sciences, New York Institute of Technology College of Osteopathic Medicine, NY 11568, USA
| | - Olivier Thibault
- Department of Pharmacological and Nutritional Sciences, College of Medicine, University of Kentucky, KY 40536, USA
| | - Ramon C. Sun
- Department of Neuroscience, College of Medicine, University of Kentucky, KY 40536, USA
- Markey Cancer Center, Lexington, KY 40536, USA
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY 40536, USA
- Department of Biochemistry and Molecular Biology, University of Florida, College of Medicine, Gainesville, FL 32611, USA
- Center for Advanced Spatial Biomolecule Research, University of Florida, College of Medicine, Gainesville, FL 32611, USA
| | - Douglas A. Andres
- Department of Molecular and Cellular Biochemistry, College of Medicine, University of Kentucky, KY 40536, USA
- Markey Cancer Center, Lexington, KY 40536, USA
- Spinal Cord and Brain Injury Research Center, College of Medicine, University of Kentucky, KY 40536, USA
- Gill Heart and Vascular Institute, College of Medicine, University of Kentucky, Lexington, KY 40536, USA
| |
Collapse
|
14
|
Wang Y, Zhang S, Lan Z, Doan V, Kim B, Liu S, Zhu M, Hull VL, Rihani S, Zhang CL, Gray JA, Guo F. SOX2 is essential for astrocyte maturation and its deletion leads to hyperactive behavior in mice. Cell Rep 2022; 41:111842. [PMID: 36543123 PMCID: PMC9875714 DOI: 10.1016/j.celrep.2022.111842] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2021] [Revised: 09/23/2022] [Accepted: 11/24/2022] [Indexed: 12/24/2022] Open
Abstract
Children with SOX2 deficiency develop ocular disorders and extra-ocular CNS anomalies. Animal data show that SOX2 is essential for retinal and neural stem cell development. In the CNS parenchyma, SOX2 is primarily expressed in astroglial and oligodendroglial cells. Here, we report a crucial role of astroglial SOX2 in postnatal brain development. Astroglial Sox2-deficient mice develop hyperactivity in locomotion and increased neuronal excitability in the corticostriatal circuit. Sox2 deficiency inhibits postnatal astrocyte maturation molecularly, morphologically, and electrophysiologically without affecting astroglia proliferation. Mechanistically, SOX2 directly binds to a cohort of astrocytic signature and functional genes, the expression of which is significantly reduced in Sox2-deficient CNS and astrocytes. Consistently, Sox2 deficiency remarkably reduces glutamate transporter expression and compromised astrocyte function of glutamate uptake. Our study provides insights into the cellular mechanisms underlying brain defects in children with SOX2 mutations and suggests a link of astrocyte SOX2 with extra-ocular abnormalities in SOX2-mutant subjects.
Collapse
Affiliation(s)
- Yan Wang
- Institute for Pediatric Regenerative Medicine, Shriners Hospitals for Children Northern California, Sacramento, CA 95817, USA; Department of Neurology, School of Medicine, University of California, Davis, Davis, CA 95817, USA
| | - Sheng Zhang
- Institute for Pediatric Regenerative Medicine, Shriners Hospitals for Children Northern California, Sacramento, CA 95817, USA; Department of Neurology, School of Medicine, University of California, Davis, Davis, CA 95817, USA
| | - Zhaohui Lan
- Institute for Pediatric Regenerative Medicine, Shriners Hospitals for Children Northern California, Sacramento, CA 95817, USA; Department of Neurology, School of Medicine, University of California, Davis, Davis, CA 95817, USA
| | - Vui Doan
- Institute for Pediatric Regenerative Medicine, Shriners Hospitals for Children Northern California, Sacramento, CA 95817, USA
| | - Bokyung Kim
- Institute for Pediatric Regenerative Medicine, Shriners Hospitals for Children Northern California, Sacramento, CA 95817, USA; Department of Neurology, School of Medicine, University of California, Davis, Davis, CA 95817, USA
| | - Sihan Liu
- Institute for Pediatric Regenerative Medicine, Shriners Hospitals for Children Northern California, Sacramento, CA 95817, USA; Department of Neurology, School of Medicine, University of California, Davis, Davis, CA 95817, USA
| | - Meina Zhu
- Institute for Pediatric Regenerative Medicine, Shriners Hospitals for Children Northern California, Sacramento, CA 95817, USA; Department of Neurology, School of Medicine, University of California, Davis, Davis, CA 95817, USA
| | - Vanessa L Hull
- Institute for Pediatric Regenerative Medicine, Shriners Hospitals for Children Northern California, Sacramento, CA 95817, USA; Department of Neurology, School of Medicine, University of California, Davis, Davis, CA 95817, USA
| | - Sami Rihani
- Institute for Pediatric Regenerative Medicine, Shriners Hospitals for Children Northern California, Sacramento, CA 95817, USA; Department of Neurology, School of Medicine, University of California, Davis, Davis, CA 95817, USA
| | - Chun-Li Zhang
- Department of Molecular Biology and Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - John A Gray
- Department of Neurology, School of Medicine, University of California, Davis, Davis, CA 95817, USA; Center for Neuroscience, University of California, Davis, Davis, CA 95618, USA
| | - Fuzheng Guo
- Institute for Pediatric Regenerative Medicine, Shriners Hospitals for Children Northern California, Sacramento, CA 95817, USA; Department of Neurology, School of Medicine, University of California, Davis, Davis, CA 95817, USA.
| |
Collapse
|
15
|
Qin C, Wen M. miR-145 from Bone Marrow Mesenchymal Stem Cells (BMSC) Improves Cardiac Function After Myocardial Infarction in Rat with Diabetes. J BIOMATER TISS ENG 2022. [DOI: 10.1166/jbt.2022.3191] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
This study assesses the role of miR-145 from BMSC in the cardiac function after MI in rat with diabetes. Rat with T1DM model was established and then were treated with PBS, DM-BMSC with overexpression of miR-145, BMSC with the knockdown of miR-145 respectively after twenty-four hours
followed by analysis of the remodeling of vessels and protein, mRNA expressions. miR-145 in DM-BMSC was significantly reduced compared with control group and DM-BMSC prolonged the survival rate of rats. The formation of blood capillary and axon growth in DM-BMSC was increased and decreased
in BMSC with knockdown of miR-145. The therapeutic action of DM-BMSC could be improved notably and remodeling of vessels and protein was increased. Smad1 was a target gene of miR-145. In conclusion, cardiac function and neurological recovery in MI is improved by miR-145 through targeting Smad1
expression, indicating that miR-145 might be a novel target for the treatment of MI.
Collapse
Affiliation(s)
- Chuanyu Qin
- Department of Cardiology, The Second Affiliated Hospital of Qiqihar Medical College, Qiqihar City, 161099, Heilongjiang Province, China
| | - Mingli Wen
- Department of Respiratory Medicine, The First Affiliated Hospital of Qiqihar Medical College, Qiqihar City, 161041, Heilongjiang Province, China
| |
Collapse
|
16
|
Shi B, Zhao J, Xu Z, Chen C, Xu L, Xu C, Sun M, Kuang H. Chiral Nanoparticles Force Neural Stem Cell Differentiation to Alleviate Alzheimer's Disease. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2202475. [PMID: 36008133 PMCID: PMC9561871 DOI: 10.1002/advs.202202475] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Revised: 08/08/2022] [Indexed: 06/04/2023]
Abstract
The differentiation of neural stem cells via nanomaterials has attracted attention and has become a potential tool. However, the chirality effect in neural stem cell differentiation has not been investigated. Here, this study shows that chiral nanoparticles (NPs) with strong chirality can efficiently accelerate the differentiation of mouse neural stem cells (NSCs) into neurons under near-infrared (NIR) light illumination. L-type NPs are 1.95 times greater than D-type NPs in promoting NSCs differentiation due to their 1.47-fold endocytosis efficiency. Whole gene expression map analysis reveals that circularly polarized light illumination and chiral NPs irradiation significantly upregulate Map2, Yap1, and Taz genes, resulting in mechanical force, cytoskeleton protein action, and accelerated NSCs differentiation. In vivo experiments show that successful differentiation can further alleviate symptoms in Alzheimer's disease mice. Moreover, the clearance of L-type NPs on amyloid and hyperphosphorylated p-tau protein reachs 68.24% and 66.43%, respectively, under the synergy of NIR irradiation. The findings suggest that strong chiral nanomaterials may have advantages in guiding cell development and can be used in biomedicine.
Collapse
Affiliation(s)
- Baimei Shi
- International Joint Research Laboratory for Biointerface and BiodetectionJiangnan UniversityWuxiJiangsu214122China
- State Key Laboratory of Food Science and TechnologyJiangnan UniversityWuxiJiangsu214122China
| | - Jing Zhao
- International Joint Research Laboratory for Biointerface and BiodetectionJiangnan UniversityWuxiJiangsu214122China
- State Key Laboratory of Food Science and TechnologyJiangnan UniversityWuxiJiangsu214122China
| | - Zhuojia Xu
- International Joint Research Laboratory for Biointerface and BiodetectionJiangnan UniversityWuxiJiangsu214122China
- State Key Laboratory of Food Science and TechnologyJiangnan UniversityWuxiJiangsu214122China
| | - Chen Chen
- International Joint Research Laboratory for Biointerface and BiodetectionJiangnan UniversityWuxiJiangsu214122China
- State Key Laboratory of Food Science and TechnologyJiangnan UniversityWuxiJiangsu214122China
| | - Liguang Xu
- International Joint Research Laboratory for Biointerface and BiodetectionJiangnan UniversityWuxiJiangsu214122China
- State Key Laboratory of Food Science and TechnologyJiangnan UniversityWuxiJiangsu214122China
| | - Chuanlai Xu
- International Joint Research Laboratory for Biointerface and BiodetectionJiangnan UniversityWuxiJiangsu214122China
- State Key Laboratory of Food Science and TechnologyJiangnan UniversityWuxiJiangsu214122China
| | - Maozhong Sun
- International Joint Research Laboratory for Biointerface and BiodetectionJiangnan UniversityWuxiJiangsu214122China
- State Key Laboratory of Food Science and TechnologyJiangnan UniversityWuxiJiangsu214122China
| | - Hua Kuang
- International Joint Research Laboratory for Biointerface and BiodetectionJiangnan UniversityWuxiJiangsu214122China
- State Key Laboratory of Food Science and TechnologyJiangnan UniversityWuxiJiangsu214122China
| |
Collapse
|
17
|
Sartoretti MM, Campetella CA, Lanuza GM. Dbx1 controls the development of astrocytes of the intermediate spinal cord by modulating Notch signaling. Development 2022; 149:275961. [DOI: 10.1242/dev.200750] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2022] [Accepted: 06/27/2022] [Indexed: 10/17/2022]
Abstract
ABSTRACT
Significant progress has been made in elucidating the basic principles that govern neuronal specification in the developing central nervous system. In contrast, much less is known about the origin of astrocytic diversity. Here, we demonstrate that a restricted pool of progenitors in the mouse spinal cord, expressing the transcription factor Dbx1, produces a subset of astrocytes, in addition to interneurons. Ventral p0-derived astrocytes (vA0 cells) exclusively populate intermediate regions of spinal cord with extraordinary precision. The postnatal vA0 population comprises gray matter protoplasmic and white matter fibrous astrocytes and a group of cells with strict radial morphology contacting the pia. We identified that vA0 cells in the lateral funiculus are distinguished by the expression of reelin and Kcnmb4. We show that Dbx1 mutants have an increased number of vA0 cells at the expense of p0-derived interneurons. Manipulation of the Notch pathway, together with the alteration in their ligands seen in Dbx1 knockouts, suggest that Dbx1 controls neuron-glial balance by modulating Notch-dependent cell interactions. In summary, this study highlights that restricted progenitors in the dorsal-ventral neural tube produce region-specific astrocytic subgroups and that progenitor transcriptional programs highly influence glial fate and are instrumental in creating astrocyte diversity.
Collapse
Affiliation(s)
- Maria Micaela Sartoretti
- Developmental Neurobiology Lab, Fundación Instituto Leloir and Consejo Nacional de Investigaciones Científicas y Técnicas (IIBBA-CONICET) , Avenida Patricias Argentinas 435, Buenos Aires 1405 , Argentina
| | - Carla A. Campetella
- Developmental Neurobiology Lab, Fundación Instituto Leloir and Consejo Nacional de Investigaciones Científicas y Técnicas (IIBBA-CONICET) , Avenida Patricias Argentinas 435, Buenos Aires 1405 , Argentina
| | - Guillermo M. Lanuza
- Developmental Neurobiology Lab, Fundación Instituto Leloir and Consejo Nacional de Investigaciones Científicas y Técnicas (IIBBA-CONICET) , Avenida Patricias Argentinas 435, Buenos Aires 1405 , Argentina
| |
Collapse
|
18
|
Dermitzakis I, Manthou ME, Meditskou S, Miliaras D, Kesidou E, Boziki M, Petratos S, Grigoriadis N, Theotokis P. Developmental Cues and Molecular Drivers in Myelinogenesis: Revisiting Early Life to Re-Evaluate the Integrity of CNS Myelin. Curr Issues Mol Biol 2022; 44:3208-3237. [PMID: 35877446 PMCID: PMC9324160 DOI: 10.3390/cimb44070222] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 07/14/2022] [Accepted: 07/17/2022] [Indexed: 02/07/2023] Open
Abstract
The mammalian central nervous system (CNS) coordinates its communication through saltatory conduction, facilitated by myelin-forming oligodendrocytes (OLs). Despite the fact that neurogenesis from stem cell niches has caught the majority of attention in recent years, oligodendrogenesis and, more specifically, the molecular underpinnings behind OL-dependent myelinogenesis, remain largely unknown. In this comprehensive review, we determine the developmental cues and molecular drivers which regulate normal myelination both at the prenatal and postnatal periods. We have indexed the individual stages of myelinogenesis sequentially; from the initiation of oligodendrocyte precursor cells, including migration and proliferation, to first contact with the axon that enlists positive and negative regulators for myelination, until the ultimate maintenance of the axon ensheathment and myelin growth. Here, we highlight multiple developmental pathways that are key to successful myelin formation and define the molecular pathways that can potentially be targets for pharmacological interventions in a variety of neurological disorders that exhibit demyelination.
Collapse
Affiliation(s)
- Iasonas Dermitzakis
- Department of Histology-Embryology, School of Medicine, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece; (I.D.); (M.E.M.); (S.M.); (D.M.)
| | - Maria Eleni Manthou
- Department of Histology-Embryology, School of Medicine, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece; (I.D.); (M.E.M.); (S.M.); (D.M.)
| | - Soultana Meditskou
- Department of Histology-Embryology, School of Medicine, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece; (I.D.); (M.E.M.); (S.M.); (D.M.)
| | - Dimosthenis Miliaras
- Department of Histology-Embryology, School of Medicine, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece; (I.D.); (M.E.M.); (S.M.); (D.M.)
| | - Evangelia Kesidou
- Laboratory of Experimental Neurology and Neuroimmunology, Second Department of Neurology, AHEPA University Hospital, 54621 Thessaloniki, Greece; (E.K.); (M.B.); (N.G.)
| | - Marina Boziki
- Laboratory of Experimental Neurology and Neuroimmunology, Second Department of Neurology, AHEPA University Hospital, 54621 Thessaloniki, Greece; (E.K.); (M.B.); (N.G.)
| | - Steven Petratos
- Department of Neuroscience, Central Clinical School, Monash University, Prahran, VIC 3004, Australia;
| | - Nikolaos Grigoriadis
- Laboratory of Experimental Neurology and Neuroimmunology, Second Department of Neurology, AHEPA University Hospital, 54621 Thessaloniki, Greece; (E.K.); (M.B.); (N.G.)
| | - Paschalis Theotokis
- Department of Histology-Embryology, School of Medicine, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece; (I.D.); (M.E.M.); (S.M.); (D.M.)
- Laboratory of Experimental Neurology and Neuroimmunology, Second Department of Neurology, AHEPA University Hospital, 54621 Thessaloniki, Greece; (E.K.); (M.B.); (N.G.)
- Correspondence:
| |
Collapse
|
19
|
Wang S, Wang Y, Zou S. A Glance at the Molecules That Regulate Oligodendrocyte Myelination. Curr Issues Mol Biol 2022; 44:2194-2216. [PMID: 35678678 PMCID: PMC9164040 DOI: 10.3390/cimb44050149] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Revised: 05/10/2022] [Accepted: 05/13/2022] [Indexed: 11/16/2022] Open
Abstract
Oligodendrocyte (OL) myelination is a critical process for the neuronal axon function in the central nervous system. After demyelination occurs because of pathophysiology, remyelination makes repairs similar to myelination. Proliferation and differentiation are the two main stages in OL myelination, and most factors commonly play converse roles in these two stages, except for a few factors and signaling pathways, such as OLIG2 (Oligodendrocyte transcription factor 2). Moreover, some OL maturation gene mutations induce hypomyelination or hypermyelination without an obvious function in proliferation and differentiation. Herein, three types of factors regulating myelination are reviewed in sequence.
Collapse
Affiliation(s)
- Shunqi Wang
- Institute of Life Science & School of Life Sciences, Nanchang University, Nanchang 330031, China; (S.W.); (Y.W.)
- School of Basic Medical Sciences, Nanchang University, Nanchang 330031, China
| | - Yingxing Wang
- Institute of Life Science & School of Life Sciences, Nanchang University, Nanchang 330031, China; (S.W.); (Y.W.)
| | - Suqi Zou
- Institute of Life Science & School of Life Sciences, Nanchang University, Nanchang 330031, China; (S.W.); (Y.W.)
- School of Basic Medical Sciences, Nanchang University, Nanchang 330031, China
- Correspondence:
| |
Collapse
|
20
|
Pouremamali F, Vahedian V, Hassani N, Mirzaei S, Pouremamali A, Kazemzadeh H, Faridvand Y, Jafari-gharabaghlou D, Nouri M, Maroufi NF. The role of SOX family in cancer stem cell maintenance: With a focus on SOX2. Pathol Res Pract 2022; 231:153783. [DOI: 10.1016/j.prp.2022.153783] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Revised: 01/17/2022] [Accepted: 01/25/2022] [Indexed: 02/06/2023]
|
21
|
Aberle T, Piefke S, Hillgärtner S, Tamm ER, Wegner M, Küspert M. OUP accepted manuscript. Nucleic Acids Res 2022; 50:1951-1968. [PMID: 35137157 PMCID: PMC8887482 DOI: 10.1093/nar/gkac042] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Revised: 12/21/2021] [Accepted: 01/14/2022] [Indexed: 11/14/2022] Open
Abstract
In oligodendrocytes of the vertebrate central nervous system a complex network of transcriptional regulators is required to ensure correct and timely myelination of neuronal axons. Here we identify Zfp276, the only mammalian ZAD-domain containing zinc finger protein, as a transcriptional regulator of oligodendrocyte differentiation and central myelination downstream of Sox10. In the central nervous system, Zfp276 is exclusively expressed in mature oligodendrocytes. Oligodendroglial deletion of Zfp276 led to strongly reduced expression of myelin genes in the early postnatal mouse spinal cord. Retroviral overexpression of Zfp276 in cultured oligodendrocyte precursor cells induced precocious expression of maturation markers and myelin genes, further supporting its role in oligodendroglial differentiation. On the molecular level, Zfp276 directly binds to and represses Sox10-dependent gene regulatory regions of immaturity factors and functionally interacts with the transcriptional repressor Zeb2 to enable fast transition of oligodendrocytes to the myelinating stage.
Collapse
Affiliation(s)
- Tim Aberle
- Institut für Biochemie, Friedrich-Alexander-Universität Erlangen-Nürnberg, D-91054, Erlangen, Germany
| | - Sandra Piefke
- Institut für Biochemie, Friedrich-Alexander-Universität Erlangen-Nürnberg, D-91054, Erlangen, Germany
| | - Simone Hillgärtner
- Institut für Biochemie, Friedrich-Alexander-Universität Erlangen-Nürnberg, D-91054, Erlangen, Germany
| | - Ernst R Tamm
- Institut für Humananatomie und Embryologie, Universität Regensburg, D-93053, Regensburg, Germany
| | - Michael Wegner
- Institut für Biochemie, Friedrich-Alexander-Universität Erlangen-Nürnberg, D-91054, Erlangen, Germany
| | - Melanie Küspert
- To whom correspondence should be addressed. Tel: +49 9131 85 24638; Fax: +49 9131 85 22484;
| |
Collapse
|
22
|
Xu Q, Yu M, Zhou Y, Huang Z, Huang X, Xu B, Zhou K, Chen X, Xia Y, Wang X, Lu C, Han X. Effects of 2,2',4,4'-tetrabromodiphenyl ether on the development of mouse embryonic stem cells. Reprod Toxicol 2021; 106:18-24. [PMID: 34547414 DOI: 10.1016/j.reprotox.2021.09.008] [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: 03/27/2021] [Revised: 07/23/2021] [Accepted: 09/14/2021] [Indexed: 12/09/2022]
Abstract
2,2',4,4'-Tetrabromodiphenyl ether (BDE47) poses potential risks to reproduction and development, but the mechanism of its toxicity has not yet been elucidated. To explore the developmental toxicity of BDE47, mouse embryonic stem cells (mESCs), which are ideal models for testing the developmental toxicity of environmental contaminants in vitro, were exposed to BDE47 (0.04 μM, 1 μM, 25 μM, or 100 μM) for 24 h or 48 h in this study. Our results indicated that BDE47 treatment changed the morphology of mESCs, inhibited cell viability and increased apoptosis. In addition, alkaline phosphatase (AP) staining in mESCs was significantly decreased after BDE47 treatment (25 μM and 100 μM), indicating that BDE47 treatment affected the pluripotency of mESCs. Through a cell immunofluorescence assay, we found that the fluorescence intensities of Oct4, Sox2 and Nanog were all significantly lower in the group treated with the highest BDE47 concentration (100 μM) than in the control group, consistent with the qRT-PCR and Western blot results. The levels of miR-145 and miR-34a, which regulate genes related to cell differentiation, were significantly increased in BDE47-treated mESCs, further clarifying the potential mechanism. Overall, our findings demonstrate that BDE47 exposure upregulates the expression of miR-145 and miR-34a and in turn downregulates the expression of Oct4, Sox2 and Nanog, thereby affecting apoptosis and pluripotency and causing toxicity during embryonic development.
Collapse
Affiliation(s)
- Qiaoqiao Xu
- State Key Laboratory of Reproductive Medicine, Institute of Toxicology, Nanjing Medical University, Nanjing, 210029, China; Key Laboratory of Modern Toxicology of Ministry of Education, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, 210029, China
| | - Mingming Yu
- State Key Laboratory of Reproductive Medicine, Institute of Toxicology, Nanjing Medical University, Nanjing, 210029, China; Key Laboratory of Modern Toxicology of Ministry of Education, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, 210029, China
| | - Yuxia Zhou
- Prenatal Diagnosis Center, Shandong Maternal and Child Health Hospital, Jinan, 250014, China
| | - Zhenyao Huang
- State Key Laboratory of Reproductive Medicine, Institute of Toxicology, Nanjing Medical University, Nanjing, 210029, China; Key Laboratory of Modern Toxicology of Ministry of Education, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, 210029, China
| | - Xiaomin Huang
- State Key Laboratory of Reproductive Medicine, Institute of Toxicology, Nanjing Medical University, Nanjing, 210029, China; Key Laboratory of Modern Toxicology of Ministry of Education, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, 210029, China
| | - Bo Xu
- State Key Laboratory of Reproductive Medicine, Institute of Toxicology, Nanjing Medical University, Nanjing, 210029, China; Key Laboratory of Modern Toxicology of Ministry of Education, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, 210029, China
| | - Kun Zhou
- State Key Laboratory of Reproductive Medicine, Institute of Toxicology, Nanjing Medical University, Nanjing, 210029, China; Key Laboratory of Modern Toxicology of Ministry of Education, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, 210029, China
| | - Xiaojiao Chen
- State Key Laboratory of Reproductive Medicine, Institute of Toxicology, Nanjing Medical University, Nanjing, 210029, China; Key Laboratory of Modern Toxicology of Ministry of Education, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, 210029, China
| | - Yankai Xia
- State Key Laboratory of Reproductive Medicine, Institute of Toxicology, Nanjing Medical University, Nanjing, 210029, China; Key Laboratory of Modern Toxicology of Ministry of Education, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, 210029, China
| | - Xinru Wang
- State Key Laboratory of Reproductive Medicine, Institute of Toxicology, Nanjing Medical University, Nanjing, 210029, China; Key Laboratory of Modern Toxicology of Ministry of Education, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, 210029, China
| | - Chuncheng Lu
- State Key Laboratory of Reproductive Medicine, Institute of Toxicology, Nanjing Medical University, Nanjing, 210029, China; Key Laboratory of Modern Toxicology of Ministry of Education, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, 210029, China
| | - Xiumei Han
- State Key Laboratory of Reproductive Medicine, Institute of Toxicology, Nanjing Medical University, Nanjing, 210029, China; Key Laboratory of Modern Toxicology of Ministry of Education, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, 210029, China.
| |
Collapse
|
23
|
Kang M, Yao Y. Laminin regulates oligodendrocyte development and myelination. Glia 2021; 70:414-429. [PMID: 34773273 DOI: 10.1002/glia.24117] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Revised: 10/26/2021] [Accepted: 10/29/2021] [Indexed: 11/08/2022]
Abstract
Oligodendrocytes are the cells that myelinate axons and provide trophic support to neurons in the CNS. Their dysfunction has been associated with a group of disorders known as demyelinating diseases, such as multiple sclerosis. Oligodendrocytes are derived from oligodendrocyte precursor cells, which differentiate into premyelinating oligodendrocytes and eventually mature oligodendrocytes. The development and function of oligodendrocytes are tightly regulated by a variety of molecules, including laminin, a major protein of the extracellular matrix. Accumulating evidence suggests that laminin actively regulates every aspect of oligodendrocyte biology, including survival, migration, proliferation, differentiation, and myelination. How can laminin exert such diverse functions in oligodendrocytes? It is speculated that the distinct laminin isoforms, laminin receptors, and/or key signaling molecules expressed in oligodendrocytes at different developmental stages are the reasons. Understanding molecular targets and signaling pathways unique to each aspect of oligodendrocyte biology will enable more accurate manipulation of oligodendrocyte development and function, which may have implications in the therapies of demyelinating diseases. Here in this review, we first introduce oligodendrocyte biology, followed by the expression of laminin and laminin receptors in oligodendrocytes and other CNS cells. Next, the functions of laminin in oligodendrocyte biology, including survival, migration, proliferation, differentiation, and myelination, are discussed in detail. Last, key questions and challenges in the field are discussed. By providing a comprehensive review on laminin's roles in OL lineage cells, we hope to stimulate novel hypotheses and encourage new research in the field.
Collapse
Affiliation(s)
- Minkyung Kang
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, Tampa, Florida, USA
| | - Yao Yao
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, Tampa, Florida, USA
| |
Collapse
|
24
|
Chang J, Lurie RH, Sharma A, Bashir M, Fung CM, Dettman RW, Dizon MLV. Intrauterine growth restriction followed by oxygen support uniquely interferes with genetic regulators of myelination. eNeuro 2021; 8:ENEURO.0263-20.2021. [PMID: 34099489 PMCID: PMC8266217 DOI: 10.1523/eneuro.0263-20.2021] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Revised: 03/23/2021] [Accepted: 04/23/2021] [Indexed: 12/24/2022] Open
Abstract
Intrauterine growth restriction (IUGR) and oxygen exposure in isolation and combination adversely affect the developing brain, putting infants at risk for neurodevelopmental disability including cerebral palsy. Rodent models of IUGR and postnatal hyperoxia have demonstrated oligodendroglial injury with subsequent white matter injury (WMI) and motor dysfunction. Here we investigate transcriptomic dysregulation in IUGR with and without hyperoxia exposure to account for the abnormal brain structure and function previously documented. We performed RNA sequencing and analysis using a mouse model of IUGR and found that IUGR, hyperoxia, and the combination of IUGR with hyperoxia (IUGR/hyperoxia) produced distinct changes in gene expression. IUGR in isolation demonstrated the fewest differentially expressed genes compared to control. In contrast, we detected several gene alterations in IUGR/hyperoxia; genes involved in myelination were strikingly downregulated. We also identified changes to specific regulators including TCF7L2, BDNF, SOX2, and DGCR8, through Ingenuity Pathway Analysis, that may contribute to impaired myelination in IUGR/hyperoxia. Our findings show that IUGR with hyperoxia induces unique transcriptional changes in the developing brain. These indicate mechanisms for increased risk for WMI in IUGR infants exposed to oxygen and suggest potential therapeutic targets to improve motor outcomes.Significance StatementThis study demonstrates that perinatal exposures of IUGR and/or postnatal hyperoxia result in distinct transcriptomic changes in the developing brain. In particular, we found that genes involved in normal developmental myelination, myelin maintenance, and remyelination were most dysregulated when IUGR was combined with hyperoxia. Understanding how multiple risk factors lead to WMI is the first step in developing future therapeutic interventions. Additionally, because oxygen exposure is often unavoidable after birth, an understanding of gene perturbations in this setting will increase our awareness of the need for tight control of oxygen use to minimize future motor disability.
Collapse
Affiliation(s)
- Jill Chang
- Children's Hospital of Chicago, Department of Pediatrics, Division of Neonatology, Chicago, Illinois, USA
| | - Robert H Lurie
- Children's Hospital of Chicago, Department of Pediatrics, Division of Neonatology, Chicago, Illinois, USA
| | - Abhineet Sharma
- Children's Hospital of Chicago, Department of Pediatrics, Division of Neonatology, Chicago, Illinois, USA
| | - Mirrah Bashir
- Children's Hospital of Chicago, Department of Pediatrics, Division of Neonatology, Chicago, Illinois, USA
| | - Camille M Fung
- University of Utah, Department of Pediatrics, Salt Lake City, Utah, USA
| | - Robert W Dettman
- Children's Hospital of Chicago, Department of Pediatrics, Division of Neonatology, Chicago, Illinois, USA
| | - Maria L V Dizon
- Children's Hospital of Chicago, Department of Pediatrics, Division of Neonatology, Chicago, Illinois, USA
| |
Collapse
|
25
|
Stevanovic M, Drakulic D, Lazic A, Ninkovic DS, Schwirtlich M, Mojsin M. SOX Transcription Factors as Important Regulators of Neuronal and Glial Differentiation During Nervous System Development and Adult Neurogenesis. Front Mol Neurosci 2021; 14:654031. [PMID: 33867936 PMCID: PMC8044450 DOI: 10.3389/fnmol.2021.654031] [Citation(s) in RCA: 78] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Accepted: 03/11/2021] [Indexed: 12/11/2022] Open
Abstract
The SOX proteins belong to the superfamily of transcription factors (TFs) that display properties of both classical TFs and architectural components of chromatin. Since the cloning of the Sox/SOX genes, remarkable progress has been made in illuminating their roles as key players in the regulation of multiple developmental and physiological processes. SOX TFs govern diverse cellular processes during development, such as maintaining the pluripotency of stem cells, cell proliferation, cell fate decisions/germ layer formation as well as terminal cell differentiation into tissues and organs. However, their roles are not limited to development since SOX proteins influence survival, regeneration, cell death and control homeostasis in adult tissues. This review summarized current knowledge of the roles of SOX proteins in control of central nervous system development. Some SOX TFs suspend neural progenitors in proliferative, stem-like state and prevent their differentiation. SOX proteins function as pioneer factors that occupy silenced target genes and keep them in a poised state for activation at subsequent stages of differentiation. At appropriate stage of development, SOX members that maintain stemness are down-regulated in cells that are competent to differentiate, while other SOX members take over their functions and govern the process of differentiation. Distinct SOX members determine down-stream processes of neuronal and glial differentiation. Thus, sequentially acting SOX TFs orchestrate neural lineage development defining neuronal and glial phenotypes. In line with their crucial roles in the nervous system development, deregulation of specific SOX proteins activities is associated with neurodevelopmental disorders (NDDs). The overview of the current knowledge about the link between SOX gene variants and NDDs is presented. We outline the roles of SOX TFs in adult neurogenesis and brain homeostasis and discuss whether impaired adult neurogenesis, detected in neurodegenerative diseases, could be associated with deregulation of SOX proteins activities. We present the current data regarding the interaction between SOX proteins and signaling pathways and microRNAs that play roles in nervous system development. Finally, future research directions that will improve the knowledge about distinct and various roles of SOX TFs in health and diseases are presented and discussed.
Collapse
Affiliation(s)
- Milena Stevanovic
- Laboratory for Human Molecular Genetics, Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, Belgrade, Serbia.,Faculty of Biology, University of Belgrade, Belgrade, Serbia.,Serbian Academy of Sciences and Arts, Belgrade, Serbia
| | - Danijela Drakulic
- Laboratory for Human Molecular Genetics, Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, Belgrade, Serbia
| | - Andrijana Lazic
- Laboratory for Human Molecular Genetics, Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, Belgrade, Serbia
| | - Danijela Stanisavljevic Ninkovic
- Laboratory for Human Molecular Genetics, Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, Belgrade, Serbia
| | - Marija Schwirtlich
- Laboratory for Human Molecular Genetics, Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, Belgrade, Serbia
| | - Marija Mojsin
- Laboratory for Human Molecular Genetics, Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, Belgrade, Serbia
| |
Collapse
|
26
|
One-step Reprogramming of Human Fibroblasts into Oligodendrocyte-like Cells by SOX10, OLIG2, and NKX6.2. Stem Cell Reports 2021; 16:771-783. [PMID: 33770499 PMCID: PMC8072064 DOI: 10.1016/j.stemcr.2021.03.001] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2020] [Revised: 03/01/2021] [Accepted: 03/01/2021] [Indexed: 01/31/2023] Open
Abstract
Limited access to human oligodendrocytes impairs better understanding of oligodendrocyte pathology in myelin diseases. Here, we describe a method to robustly convert human fibroblasts directly into oligodendrocyte-like cells (dc-hiOLs), which allows evaluation of remyelination-promoting compounds and disease modeling. Ectopic expression of SOX10, OLIG2, and NKX6.2 in human fibroblasts results in rapid generation of O4+ cells, which further differentiate into MBP+ mature oligodendrocyte-like cells within 16 days. dc-hiOLs undergo chromatin remodeling to express oligodendrocyte markers, ensheath axons, and nanofibers in vitro, respond to promyelination compound treatment, and recapitulate in vitro oligodendroglial pathologies associated with Pelizaeus-Merzbacher leukodystrophy related to PLP1 mutations. Furthermore, DNA methylome analysis provides evidence that the CpG methylation pattern significantly differs between dc-hiOLs derived from fibroblasts of young and old donors, indicating the maintenance of the source cells’ “age.” In summary, dc-hiOLs represent a reproducible technology that could contribute to personalized medicine in the field of myelin diseases. SOX10, OLIG2, and NKX6.2 directly convert human fibroblasts into dc-hiOLs in 16 days dc-hiOLs express key oligodendrocyte markers dc-hiOLs preserve the epigenetic age of donor cells dc-hiOLs from PMD patients show maturation deficit and vulnerability to cell death
Collapse
|
27
|
Xu R, Shen X, Xie H, Zhang H, Liu D, Chen X, Fu Y, Zhang P, Yang Y, Cheng J, Jiang H. Identification of the canonical and noncanonical role of miR-143/145 in estrogen-deficient bone loss. Theranostics 2021; 11:5491-5510. [PMID: 33859759 PMCID: PMC8039936 DOI: 10.7150/thno.55041] [Citation(s) in RCA: 4] [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: 10/26/2020] [Accepted: 02/24/2021] [Indexed: 12/16/2022] Open
Abstract
Rationale: Postmenopausal-induced bone loss is mainly caused by declining core transcription factors (TFs) of bone mesenchymal stem cells (BMSCs), but little is known about how miRNAs regulate chromatin structure remodeling of TFs gene to maintain BMSCs function in bone homeostasis. Methods: We examined the serum, salivary and bone samples from Pre- and Post-menopause women by paired analysis and confirmed canonical ceRNA role of MIR143HG and miR-143/145 complexes in cytoplasm and noncanonical role for SOX2 transcription in nucleus (FISH, qRT-PCR, immunostaining, Luciferase assays and ChIP). Moreover, we took advantage of transgenic mice under OVX-induced osteoporosis, studying the in vitro and in vivo effect of miR-143/145 deletion on BMSCs function and bone homeostasis. Last, using miRNA antagonism, antagomiR-143/145 were delivered into bone marrow to treat estrogen-deficient bone loss. Results: Here, we identified miR-143/145 as potential diagnostic candidates for postmenopausal osteoporosis, and miR-143/145 overexpression impaired BMSCs self-renewing and differentiation function. Mechanistically, we confirmed that cytoplasmic miR-143/145 and LncRNA MIR143HG, that controlled by ERβ, cooperatively regulated pluripotency genes translation via canonical ceRNA pathway, and MIR143HG cooperates with miR‑143 to nuclear translocation for co-activation of SOX2 transcription via opening promoter chromatin. Meanwhile, miR‑143/145 were shuttled into osteoclasts in extracellular vesicles and triggered osteoclastic activity by targeting Cd226 and Srgap2. Furthermore, miR-143/145-/- mice or using chemically‑modified antagomiR-143/145 significantly alleviated estrogen-deficient osteoporosis. Conclusions: Our findings reveal a canonical and noncanonical role of miR-143/145 in controlling BMSCs pluripotency and unfold their dual effect on bone formation and bone resorption, suggesting miR-143/145 as promising therapeutic targets for treating estrogen-deficient bone loss.
Collapse
Affiliation(s)
- Rongyao Xu
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing 210029, Jiangsu Province, China
- Department of Oral and Maxillofacial Surgery, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing 210029, Jiangsu Province, China
| | - Xin Shen
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing 210029, Jiangsu Province, China
- Department of Oral and Maxillofacial Surgery, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing 210029, Jiangsu Province, China
| | - Hanyu Xie
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing 210029, Jiangsu Province, China
- Department of Oral and Maxillofacial Surgery, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing 210029, Jiangsu Province, China
| | - Hengguo Zhang
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing 210029, Jiangsu Province, China
- Department of Oral and Maxillofacial Surgery, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing 210029, Jiangsu Province, China
| | - Dingshan Liu
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing 210029, Jiangsu Province, China
- Department of Oral and Maxillofacial Surgery, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing 210029, Jiangsu Province, China
| | - Xin Chen
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing 210029, Jiangsu Province, China
- Department of Oral and Maxillofacial Surgery, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing 210029, Jiangsu Province, China
| | - Yu Fu
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing 210029, Jiangsu Province, China
- Department of Oral and Maxillofacial Surgery, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing 210029, Jiangsu Province, China
| | - Ping Zhang
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing 210029, Jiangsu Province, China
- Department of Oral and Maxillofacial Surgery, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing 210029, Jiangsu Province, China
| | - Yi Yang
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing 210029, Jiangsu Province, China
- Department of Dental Implantology, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing 210029, Jiangsu Province, China
| | - Jie Cheng
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing 210029, Jiangsu Province, China
- Department of Oral and Maxillofacial Surgery, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing 210029, Jiangsu Province, China
| | - Hongbing Jiang
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing 210029, Jiangsu Province, China
- Department of Oral and Maxillofacial Surgery, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing 210029, Jiangsu Province, China
| |
Collapse
|
28
|
Cellular senescence and failure of myelin repair in multiple sclerosis. Mech Ageing Dev 2020; 192:111366. [DOI: 10.1016/j.mad.2020.111366] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Revised: 08/10/2020] [Accepted: 09/23/2020] [Indexed: 01/10/2023]
|
29
|
Pooyan P, Karamzadeh R, Mirzaei M, Meyfour A, Amirkhan A, Wu Y, Gupta V, Baharvand H, Javan M, Salekdeh GH. The Dynamic Proteome of Oligodendrocyte Lineage Differentiation Features Planar Cell Polarity and Macroautophagy Pathways. Gigascience 2020; 9:giaa116. [PMID: 33128372 PMCID: PMC7601170 DOI: 10.1093/gigascience/giaa116] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Revised: 07/22/2020] [Accepted: 09/28/2020] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Generation of oligodendrocytes is a sophisticated multistep process, the mechanistic underpinnings of which are not fully understood and demand further investigation. To systematically profile proteome dynamics during human embryonic stem cell differentiation into oligodendrocytes, we applied in-depth quantitative proteomics at different developmental stages and monitored changes in protein abundance using a multiplexed tandem mass tag-based proteomics approach. FINDINGS Our proteome data provided a comprehensive protein expression profile that highlighted specific expression clusters based on the protein abundances over the course of human oligodendrocyte lineage differentiation. We identified the eminence of the planar cell polarity signalling and autophagy (particularly macroautophagy) in the progression of oligodendrocyte lineage differentiation-the cooperation of which is assisted by 106 and 77 proteins, respectively, that showed significant expression changes in this differentiation process. Furthermore, differentially expressed protein analysis of the proteome profile of oligodendrocyte lineage cells revealed 378 proteins that were specifically upregulated only in 1 differentiation stage. In addition, comparative pairwise analysis of differentiation stages demonstrated that abundances of 352 proteins differentially changed between consecutive differentiation time points. CONCLUSIONS Our study provides a comprehensive systematic proteomics profile of oligodendrocyte lineage cells that can serve as a resource for identifying novel biomarkers from these cells and for indicating numerous proteins that may contribute to regulating the development of myelinating oligodendrocytes and other cells of oligodendrocyte lineage. We showed the importance of planar cell polarity signalling in oligodendrocyte lineage differentiation and revealed the autophagy-related proteins that participate in oligodendrocyte lineage differentiation.
Collapse
Affiliation(s)
- Paria Pooyan
- Department of Molecular Systems Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, Banihashem St., ACECR, Tehran 16635-148, Iran
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, Banihashem St., ACECR, Tehran 16635-148, Iran
- Department of Brain and Cognitive Science, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, Banihashem St., ACECR, Tehran 16635-148, Iran
| | - Razieh Karamzadeh
- Department of Molecular Systems Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, Banihashem St., ACECR, Tehran 16635-148, Iran
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, Banihashem St., ACECR, Tehran 16635-148, Iran
- Department of Brain and Cognitive Science, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, Banihashem St., ACECR, Tehran 16635-148, Iran
| | - Mehdi Mirzaei
- Department of Molecular Sciences, Macquarie University, North Ryde, Sydney, NSW 2109, Australia
- Australian Proteome Analysis Facility, Macquarie University, North Ryde, NSW 2109, Australia
| | - Anna Meyfour
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Daneshjoo Blv., Velenjak, Tehran 19839-63113, Iran
| | - Ardeshir Amirkhan
- Australian Proteome Analysis Facility, Macquarie University, North Ryde, NSW 2109, Australia
| | - Yunqi Wu
- Australian Proteome Analysis Facility, Macquarie University, North Ryde, NSW 2109, Australia
| | - Vivek Gupta
- Department of Clinical Medicine, Macquarie University, North Ryde, Sydney, NSW 2109, Australia
| | - Hossein Baharvand
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, Banihashem St., ACECR, Tehran 16635-148, Iran
- Department of Brain and Cognitive Science, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, Banihashem St., ACECR, Tehran 16635-148, Iran
- Department of Developmental Biology, University of Science and Culture, Ashrafi Esfahani, Tehran 1461968151, Iran
| | - Mohammad Javan
- Department of Brain and Cognitive Science, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, Banihashem St., ACECR, Tehran 16635-148, Iran
- Department of Physiology, Faculty of Medical Sciences, Tarbiat Modares University, Jalal AleAhmad, Tehran 14115-111, Iran
| | - Ghasem Hosseini Salekdeh
- Department of Molecular Systems Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, Banihashem St., ACECR, Tehran 16635-148, Iran
- Department of Molecular Sciences, Macquarie University, North Ryde, Sydney, NSW 2109, Australia
| |
Collapse
|
30
|
Wüst HM, Wegener A, Fröb F, Hartwig AC, Wegwitz F, Kari V, Schimmel M, Tamm ER, Johnsen SA, Wegner M, Sock E. Egr2-guided histone H2B monoubiquitination is required for peripheral nervous system myelination. Nucleic Acids Res 2020; 48:8959-8976. [PMID: 32672815 PMCID: PMC7498331 DOI: 10.1093/nar/gkaa606] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Revised: 07/03/2020] [Accepted: 07/07/2020] [Indexed: 12/30/2022] Open
Abstract
Schwann cells are the nerve ensheathing cells of the peripheral nervous system. Absence, loss and malfunction of Schwann cells or their myelin sheaths lead to peripheral neuropathies such as Charcot-Marie-Tooth disease in humans. During Schwann cell development and myelination chromatin is dramatically modified. However, impact and functional relevance of these modifications are poorly understood. Here, we analyzed histone H2B monoubiquitination as one such chromatin modification by conditionally deleting the Rnf40 subunit of the responsible E3 ligase in mice. Rnf40-deficient Schwann cells were arrested immediately before myelination or generated abnormally thin, unstable myelin, resulting in a peripheral neuropathy characterized by hypomyelination and progressive axonal degeneration. By combining sequencing techniques with functional studies we show that H2B monoubiquitination does not influence global gene expression patterns, but instead ensures selective high expression of myelin and lipid biosynthesis genes and proper repression of immaturity genes. This requires the specific recruitment of the Rnf40-containing E3 ligase by Egr2, the central transcriptional regulator of peripheral myelination, to its target genes. Our study identifies histone ubiquitination as essential for Schwann cell myelination and unravels new disease-relevant links between chromatin modifications and transcription factors in the underlying regulatory network.
Collapse
Affiliation(s)
- Hannah M Wüst
- Institut für Biochemie, Emil-Fischer-Zentrum, Friedrich-Alexander-Universität Erlangen-Nürnberg, Fahrstrasse 17, D-91054 Erlangen, Germany
| | - Amélie Wegener
- Institut für Biochemie, Emil-Fischer-Zentrum, Friedrich-Alexander-Universität Erlangen-Nürnberg, Fahrstrasse 17, D-91054 Erlangen, Germany
| | - Franziska Fröb
- Institut für Biochemie, Emil-Fischer-Zentrum, Friedrich-Alexander-Universität Erlangen-Nürnberg, Fahrstrasse 17, D-91054 Erlangen, Germany
| | - Anna C Hartwig
- Institut für Biochemie, Emil-Fischer-Zentrum, Friedrich-Alexander-Universität Erlangen-Nürnberg, Fahrstrasse 17, D-91054 Erlangen, Germany
| | - Florian Wegwitz
- Department of General, Visceral, and Pediatric Surgery, University Medical Center Göttingen, D-37075 Göttingen, Germany
| | - Vijayalakshmi Kari
- Department of General, Visceral, and Pediatric Surgery, University Medical Center Göttingen, D-37075 Göttingen, Germany
| | - Margit Schimmel
- Institut für Humananatomie und Embryologie, Universität Regensburg, Universitätsstrasse 31, D-93053 Regensburg, Germany
| | - Ernst R Tamm
- Institut für Humananatomie und Embryologie, Universität Regensburg, Universitätsstrasse 31, D-93053 Regensburg, Germany
| | - Steven A Johnsen
- Department of General, Visceral, and Pediatric Surgery, University Medical Center Göttingen, D-37075 Göttingen, Germany.,Gene Regulatory Mechanisms and Molecular Epigenetics Lab, Division of Gastroenterology and Hepatology, Mayo Clinic, 200 First St SW, Rochester, MN, USA
| | - Michael Wegner
- Institut für Biochemie, Emil-Fischer-Zentrum, Friedrich-Alexander-Universität Erlangen-Nürnberg, Fahrstrasse 17, D-91054 Erlangen, Germany
| | - Elisabeth Sock
- Institut für Biochemie, Emil-Fischer-Zentrum, Friedrich-Alexander-Universität Erlangen-Nürnberg, Fahrstrasse 17, D-91054 Erlangen, Germany
| |
Collapse
|
31
|
Kornfeld SF, Cummings SE, Fathi S, Bonin SR, Kothary R. MiRNA-145-5p prevents differentiation of oligodendrocyte progenitor cells by regulating expression of myelin gene regulatory factor. J Cell Physiol 2020; 236:997-1012. [PMID: 32602617 DOI: 10.1002/jcp.29910] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Revised: 06/17/2020] [Accepted: 06/17/2020] [Indexed: 12/28/2022]
Abstract
The roles of specific microRNAs (miRNA) in oligodendrocyte (OL) differentiation have been studied in depth. However, miRNAs in OL precursors and oligodendrocyte progenitor cells (OPCs) have been less extensively investigated. MiR-145-5p is highly expressed in OPCs relative to differentiating OLs, suggesting this miRNA may serve a function specifically in OPCs. Knockdown of miR-145-5p in primary OPCs led to spontaneous differentiation, as evidenced by an increased proportion of MAG+ cells, increased cell ramification, and upregulation of multiple myelin genes including MYRF, TPPP, and MAG, and OL cell cycle exit marker Cdkn1c. Supporting this transition to a differentiating state, proliferation was reduced in miR-145-5p knockdown OPCs. Further, knockdown of miR-145-5p in differentiating OLs showed enhanced differentiation, with increased branching, myelin membrane production, and myelin gene expression. We identified several OL-specific genes targeted by miR-145-5p that exhibited upregulation with miR-145-5p knockdown, including myelin gene regulatory factor (MYRF), that could be regulating the prodifferentiation phenotype in both miR-145 knockdown OPCs and OLs. Indeed, spontaneous differentiation with knockdown of miR-145-5p was fully rescued by concurrent knockdown of MYRF. However, proliferation rate was only partially rescued with MYRF knockdown, and overexpression of miR-145-5p in OPCs increased proliferation rate without affecting expression of already lowly expressed differentiation genes. Taken together, these data suggest that in OPCs miR-145-5p both prevents differentiation at least in part by preventing expression of MYRF and promotes proliferation via as-yet-unidentified mechanisms. These findings clarify the need for differential regulation of miR-145-5p between OPCs and OLs and may have further implications in demyelinating diseases such as multiple sclerosis where miR-145-5p is dysregulated.
Collapse
Affiliation(s)
- Samantha F Kornfeld
- Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, Canada.,Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Canada
| | - Sarah E Cummings
- Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, Canada.,Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Canada
| | - Samaneh Fathi
- Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, Canada
| | - Sawyer R Bonin
- Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, Canada
| | - Rashmi Kothary
- Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, Canada.,Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Canada.,Department of Medicine, University of Ottawa, Ottawa, Canada.,Centre for Neuromuscular Disease, University of Ottawa, Ottawa, Canada
| |
Collapse
|
32
|
Gong J, Hu S, Huang Z, Hu Y, Wang X, Zhao J, Qian P, Wang C, Sheng J, Lu X, Wei G, Liu D. The Requirement of Sox2 for the Spinal Cord Motor Neuron Development of Zebrafish. Front Mol Neurosci 2020; 13:34. [PMID: 32292330 PMCID: PMC7135881 DOI: 10.3389/fnmol.2020.00034] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2019] [Accepted: 02/18/2020] [Indexed: 12/13/2022] Open
Abstract
Sex-determining region Y box 2 (Sox2), expressed in neural tissues, plays an important role as a transcription factor not only in the pluripotency and proliferation of neuronal cells but also in the opposite function of cell differentiation. Nevertheless, how Sox2 is linked to motor neuron development remains unknown. Here, we showed that Sox2 was localized in the motor neurons of spinal cord by in situ hybridization and cell separation, which acted as a positive regulator of motor neuron development. The deficiency of Sox2 in zebrafish larvae resulted in abnormal PMN development, including truncated but excessively branched CaP axons, loss of MiP, and increase of undifferentiated neuron cells. Importantly, transcriptome analysis showed that Sox2-depleted embryos caused many neurogenesis, axonogenesis, axon guidance, and differentiation-related gene expression changes, which further support the vital function of Sox2 in motor neuron development. Taken together, these data indicate that Sox2 plays a crucial role in the motor neuron development by regulating neuron differentiation and morphology of neuron axons.
Collapse
Affiliation(s)
- Jie Gong
- School of Life Science, Nantong University, Nantong, China
| | - Songqun Hu
- Department of Otorhinolaryngology Head and Neck Surgery, Affiliated Hospital of Nantong University, Nantong, China
| | - Zigang Huang
- School of Life Science, Nantong University, Nantong, China
| | - Yuebo Hu
- Key Laboratory of Neuroregeneration of Jiangsu and MOE, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China
| | - Xiaoning Wang
- Key Laboratory of Neuroregeneration of Jiangsu and MOE, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China
| | - Jinxiang Zhao
- Key Laboratory of Neuroregeneration of Jiangsu and MOE, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China
| | - Peipei Qian
- Key Laboratory of Neuroregeneration of Jiangsu and MOE, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China
| | - Cheng Wang
- School of Life Science, Nantong University, Nantong, China
| | - Jiajing Sheng
- School of Life Science, Nantong University, Nantong, China
| | - Xiaofeng Lu
- School of Life Science, Nantong University, Nantong, China
| | - Guanyun Wei
- School of Life Science, Nantong University, Nantong, China
| | - Dong Liu
- School of Life Science, Nantong University, Nantong, China.,Key Laboratory of Neuroregeneration of Jiangsu and MOE, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China
| |
Collapse
|
33
|
Aprato J, Sock E, Weider M, Elsesser O, Fröb F, Wegner M. Myrf guides target gene selection of transcription factor Sox10 during oligodendroglial development. Nucleic Acids Res 2020; 48:1254-1270. [PMID: 31828317 PMCID: PMC7026603 DOI: 10.1093/nar/gkz1158] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Revised: 11/25/2019] [Accepted: 11/28/2019] [Indexed: 12/13/2022] Open
Abstract
Oligodendrocytes generate myelin in the vertebrate central nervous system and thus ensure rapid propagation of neuronal activity. Their development is controlled by a network of transcription factors that function as determinants of cell identity or as temporally restricted stage-specific regulators. The continuously expressed Sox10 and Myrf, a factor induced during late development, are particularly important for terminal differentiation. How these factors function together mechanistically and influence each other, is not well understood. Here we show that Myrf not only cooperates with Sox10 during the induction of genes required for differentiation and myelin formation. Myrf also inhibits the activity of Sox10 on genes that are essential during earlier phases of oligodendroglial development. By characterization of the exact DNA-binding requirements of Myrf, we furthermore show that cooperative activation is a consequence of joint binding of Sox10 and Myrf to the same regulatory regions. In contrast, inhibition of Sox10-dependent gene activation occurs on genes that lack Myrf binding sites and likely involves physical interaction between Myrf and Sox10 followed by sequestration. These two opposite activities allow Myrf to redirect Sox10 from genes that it activates in oligodendrocyte precursor cells to genes that need to be induced during terminal differentiation.
Collapse
Affiliation(s)
- Jessica Aprato
- Institut für Biochemie, Emil-Fischer-Zentrum, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Elisabeth Sock
- Institut für Biochemie, Emil-Fischer-Zentrum, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Matthias Weider
- Institut für Biochemie, Emil-Fischer-Zentrum, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Olga Elsesser
- Institut für Biochemie, Emil-Fischer-Zentrum, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Franziska Fröb
- Institut für Biochemie, Emil-Fischer-Zentrum, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Michael Wegner
- Institut für Biochemie, Emil-Fischer-Zentrum, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
- To whom correspondence should be addressed. Tel: +49 9131 85 24620;
| |
Collapse
|
34
|
Wittstatt J, Weider M, Wegner M, Reiprich S. MicroRNA miR‐204 regulates proliferation and differentiation of oligodendroglia in culture. Glia 2020; 68:2015-2027. [DOI: 10.1002/glia.23821] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Revised: 02/21/2020] [Accepted: 03/02/2020] [Indexed: 12/29/2022]
Affiliation(s)
- Jan Wittstatt
- Institut für Biochemie, Emil‐Fischer‐ZentrumFriedrich‐Alexander‐Universität Erlangen‐Nürnberg Erlangen Germany
| | - Matthias Weider
- Institut für Biochemie, Emil‐Fischer‐ZentrumFriedrich‐Alexander‐Universität Erlangen‐Nürnberg Erlangen Germany
| | - Michael Wegner
- Institut für Biochemie, Emil‐Fischer‐ZentrumFriedrich‐Alexander‐Universität Erlangen‐Nürnberg Erlangen Germany
| | - Simone Reiprich
- Institut für Biochemie, Emil‐Fischer‐ZentrumFriedrich‐Alexander‐Universität Erlangen‐Nürnberg Erlangen Germany
| |
Collapse
|
35
|
Chew LJ, Ming X, McEllin B, Dupree J, Hong E, Catron M, Fauveau M, Nait-Oumesmar B, Gallo V. Sox17 Regulates a Program of Oligodendrocyte Progenitor Cell Expansion and Differentiation during Development and Repair. Cell Rep 2019; 29:3173-3186.e7. [PMID: 31801081 PMCID: PMC7191642 DOI: 10.1016/j.celrep.2019.10.121] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2018] [Revised: 05/07/2019] [Accepted: 10/29/2019] [Indexed: 11/30/2022] Open
Abstract
Sox17, a SoxF family member transiently upregulated during postnatal oligodendrocyte (OL) development, promotes OL cell differentiation, but its function in white matter development and pathology in vivo is unknown. Our analysis of oligodendroglial- and OL-progenitor-cell-targeted ablation in vivo using a floxed Sox17 mouse establishes a dependence of postnatal oligodendrogenesis on Sox17 and reveals Notch signaling as a mediator of Sox17 function. Following Sox17 ablation, reduced numbers of Olig2-expressing cells and mature OLs led to developmental hypomyelination and motor dysfunction. After demyelination, Sox17 deficiency inhibited OL regeneration. OL decline was unexpectedly preceded by transiently increased differentiation and a reduction of OL progenitor cells. Evidence of a dual role for Sox17 in progenitor cell expansion by Notch and differentiation involving TCF7L2 expression were found. A program of progenitor expansion and differentiation promoted by Sox17 through Notch thus contributes to OL production and determines the outcome of white matter repair.
Collapse
Affiliation(s)
- Li-Jin Chew
- Center for Neuroscience Research, Children's National Hospital, Washington, DC 20010, USA.
| | - Xiaotian Ming
- Center for Neuroscience Research, Children's National Hospital, Washington, DC 20010, USA
| | - Brian McEllin
- Center for Neuroscience Research, Children's National Hospital, Washington, DC 20010, USA
| | - Jeffrey Dupree
- Department of Anatomy and Neurobiology, Virginia Commonwealth University Medical Center, Richmond, VA, USA; Research Service, Hunter Holmes McGuire VA Medical Center, Richmond, VA 23249, USA
| | - Elim Hong
- Center for Neuroscience Research, Children's National Hospital, Washington, DC 20010, USA
| | - Mackenzie Catron
- Center for Neuroscience Research, Children's National Hospital, Washington, DC 20010, USA
| | - Melissa Fauveau
- Institut du Cerveau et de la Moelle Épinière, ICM, INSERM U1127, CNRS UMR7225, Sorbonne Université, Hôpital de la Pitié-Salpêtrière, 75013 Paris, France
| | - Brahim Nait-Oumesmar
- Institut du Cerveau et de la Moelle Épinière, ICM, INSERM U1127, CNRS UMR7225, Sorbonne Université, Hôpital de la Pitié-Salpêtrière, 75013 Paris, France
| | - Vittorio Gallo
- Center for Neuroscience Research, Children's National Hospital, Washington, DC 20010, USA.
| |
Collapse
|
36
|
He Y, An J, Yin JJ, Sui RX, Miao Q, Ding ZB, Han QX, Wang Q, Ma CG, Xiao BG. Ethyl pyruvate enhances spontaneous remyelination by targeting microglia phagocytosis. Int Immunopharmacol 2019; 77:105929. [DOI: 10.1016/j.intimp.2019.105929] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2019] [Revised: 09/03/2019] [Accepted: 09/21/2019] [Indexed: 01/09/2023]
|
37
|
Vogel JK, Weider M, Engler LA, Hillgärtner S, Schmitt C, Hermans-Borgmeyer I, Wegner M. Sox9 overexpression exerts multiple stage-dependent effects on mouse spinal cord development. Glia 2019; 68:932-946. [PMID: 31724774 DOI: 10.1002/glia.23752] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Revised: 10/29/2019] [Accepted: 10/30/2019] [Indexed: 01/29/2023]
Abstract
The high-mobility-group (HMG)-domain protein Sox9 is one of few transcription factors implicated in gliogenesis in the vertebrate central nervous system. To further study the role of Sox9 in early spinal cord development, we generated a mouse that allows expression of Sox9 in a temporally and spatially controlled manner. Using this mouse, we show that premature Sox9 expression in neural precursor cells disrupted the neuroepithelium of the ventricular zone. Sox9 also compromised development and survival of neuronal precursors and neurons. Additionally, we observed in these mice substantial increases in oligodendroglial and astroglial cells. Reversing the normal order of appearance of essential transcriptional regulators during oligodendrogenesis, Sox10 preceded Olig2. Our study reinforces the notion that Sox9 has a strong gliogenic activity. It also argues that Sox9 expression has to be tightly controlled to prevent negative effects on early spinal cord structure and neuronal development.
Collapse
Affiliation(s)
- Julia K Vogel
- Institut für Biochemie, Emil-Fischer-Zentrum, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Matthias Weider
- Institut für Biochemie, Emil-Fischer-Zentrum, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Lisa A Engler
- Institut für Biochemie, Emil-Fischer-Zentrum, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Simone Hillgärtner
- Institut für Biochemie, Emil-Fischer-Zentrum, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Christian Schmitt
- Institut für Biochemie, Emil-Fischer-Zentrum, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | | | - Michael Wegner
- Institut für Biochemie, Emil-Fischer-Zentrum, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| |
Collapse
|
38
|
Tiane A, Schepers M, Rombaut B, Hupperts R, Prickaerts J, Hellings N, van den Hove D, Vanmierlo T. From OPC to Oligodendrocyte: An Epigenetic Journey. Cells 2019; 8:E1236. [PMID: 31614602 PMCID: PMC6830107 DOI: 10.3390/cells8101236] [Citation(s) in RCA: 68] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2019] [Revised: 10/08/2019] [Accepted: 10/10/2019] [Indexed: 12/15/2022] Open
Abstract
Oligodendrocytes provide metabolic and functional support to neuronal cells, rendering them key players in the functioning of the central nervous system. Oligodendrocytes need to be newly formed from a pool of oligodendrocyte precursor cells (OPCs). The differentiation of OPCs into mature and myelinating cells is a multistep process, tightly controlled by spatiotemporal activation and repression of specific growth and transcription factors. While oligodendrocyte turnover is rather slow under physiological conditions, a disruption in this balanced differentiation process, for example in case of a differentiation block, could have devastating consequences during ageing and in pathological conditions, such as multiple sclerosis. Over the recent years, increasing evidence has shown that epigenetic mechanisms, such as DNA methylation, histone modifications, and microRNAs, are major contributors to OPC differentiation. In this review, we discuss how these epigenetic mechanisms orchestrate and influence oligodendrocyte maturation. These insights are a crucial starting point for studies that aim to identify the contribution of epigenetics in demyelinating diseases and may thus provide new therapeutic targets to induce myelin repair in the long run.
Collapse
Affiliation(s)
- Assia Tiane
- Department of Immunology, Biomedical Research Institute, Hasselt University, Hasselt 3500, Belgium.
- Department Psychiatry and Neuropsychology, European Graduate School of Neuroscience, School for Mental Health and Neuroscience, Maastricht University, Maastricht 6200 MD, The Netherlands.
| | - Melissa Schepers
- Department of Immunology, Biomedical Research Institute, Hasselt University, Hasselt 3500, Belgium.
- Department Psychiatry and Neuropsychology, European Graduate School of Neuroscience, School for Mental Health and Neuroscience, Maastricht University, Maastricht 6200 MD, The Netherlands.
| | - Ben Rombaut
- Department of Immunology, Biomedical Research Institute, Hasselt University, Hasselt 3500, Belgium.
- Department Psychiatry and Neuropsychology, European Graduate School of Neuroscience, School for Mental Health and Neuroscience, Maastricht University, Maastricht 6200 MD, The Netherlands.
| | - Raymond Hupperts
- Department of Neurology, Zuyderland Medical Center, Sittard-Geleen 6130 MB, The Netherlands.
- Department Psychiatry and Neuropsychology, European Graduate School of Neuroscience, School for Mental Health and Neuroscience, Maastricht University, Maastricht 6200 MD, The Netherlands.
| | - Jos Prickaerts
- Department Psychiatry and Neuropsychology, European Graduate School of Neuroscience, School for Mental Health and Neuroscience, Maastricht University, Maastricht 6200 MD, The Netherlands.
| | - Niels Hellings
- Department of Immunology, Biomedical Research Institute, Hasselt University, Hasselt 3500, Belgium.
| | - Daniel van den Hove
- Department Psychiatry and Neuropsychology, European Graduate School of Neuroscience, School for Mental Health and Neuroscience, Maastricht University, Maastricht 6200 MD, The Netherlands.
- Department of Psychiatry, Psychosomatics and Psychotherapy, University of Wuerzburg, Wuerzburg 97080, Germany.
| | - Tim Vanmierlo
- Department of Immunology, Biomedical Research Institute, Hasselt University, Hasselt 3500, Belgium.
- Department Psychiatry and Neuropsychology, European Graduate School of Neuroscience, School for Mental Health and Neuroscience, Maastricht University, Maastricht 6200 MD, The Netherlands.
| |
Collapse
|
39
|
Mercurio S, Serra L, Nicolis SK. More than just Stem Cells: Functional Roles of the Transcription Factor Sox2 in Differentiated Glia and Neurons. Int J Mol Sci 2019; 20:E4540. [PMID: 31540269 PMCID: PMC6769708 DOI: 10.3390/ijms20184540] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2019] [Revised: 09/02/2019] [Accepted: 09/06/2019] [Indexed: 02/06/2023] Open
Abstract
The Sox2 transcription factor, encoded by a gene conserved in animal evolution, has become widely known because of its functional relevance for stem cells. In the developing nervous system, Sox2 is active in neural stem cells, and important for their self-renewal; differentiation to neurons and glia normally involves Sox2 downregulation. Recent evidence, however, identified specific types of fully differentiated neurons and glia that retain high Sox2 expression, and critically require Sox2 function, as revealed by functional studies in mouse and in other animals. Sox2 was found to control fundamental aspects of the biology of these cells, such as the development of correct neuronal connectivity. Sox2 downstream target genes identified within these cell types provide molecular mechanisms for cell-type-specific Sox2 neuronal and glial functions. SOX2 mutations in humans lead to a spectrum of nervous system defects, involving vision, movement control, and cognition; the identification of neurons and glia requiring Sox2 function, and the investigation of Sox2 roles and molecular targets within them, represents a novel perspective for the understanding of the pathogenesis of these defects.
Collapse
Affiliation(s)
- Sara Mercurio
- Department of Biotechnology and Biosciences, University Milano-Bicocca, 20126 Milano, Italy.
| | - Linda Serra
- Department of Biotechnology and Biosciences, University Milano-Bicocca, 20126 Milano, Italy
- CNRS, Inserm, iBV, Université Côte d'Azur, 06108 Nice, France
| | - Silvia K Nicolis
- Department of Biotechnology and Biosciences, University Milano-Bicocca, 20126 Milano, Italy.
| |
Collapse
|
40
|
Crazy Little Thing Called Sox-New Insights in Oligodendroglial Sox Protein Function. Int J Mol Sci 2019; 20:ijms20112713. [PMID: 31159496 PMCID: PMC6600536 DOI: 10.3390/ijms20112713] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Revised: 05/29/2019] [Accepted: 05/30/2019] [Indexed: 02/06/2023] Open
Abstract
In the central nervous system, oligodendrocytes wrap axons with myelin sheaths, which is essential for rapid transfer of electric signals and their trophic support. In oligodendroglia, transcription factors of the Sox protein family are pivotal regulators of a variety of developmental processes. These include specification, proliferation, and migration of oligodendrocyte precursor cells as well as terminal differentiation to mature myelinating oligodendrocytes. Sox proteins are further affected in demyelinating diseases and are involved in remyelination following damage of the central nervous system. Here we summarize and discuss latest findings on transcriptional regulation of Sox proteins, their function, target genes, and interaction with other transcription factors and chromatin remodelers in oligodendroglia with physiological and pathophysiological relevance.
Collapse
|
41
|
Sock E, Wegner M. Transcriptional control of myelination and remyelination. Glia 2019; 67:2153-2165. [PMID: 31038810 DOI: 10.1002/glia.23636] [Citation(s) in RCA: 71] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2019] [Revised: 04/01/2019] [Accepted: 04/11/2019] [Indexed: 12/11/2022]
Abstract
Myelination is an evolutionary recent differentiation program that has been independently acquired in vertebrates by Schwann cells in the peripheral nervous system and oligodendrocytes in the central nervous system. Therefore, it is not surprising that regulating transcription factors differ substantially between both cell types. However, overall principles are similar as transcriptional control in Schwann cells and oligodendrocytes combines lineage determining and stage-specific factors in complex regulatory networks. Myelination does not only occur during development, but also as remyelination in the adult. In line with the different conditions during developmental myelination and remyelination and the distinctive properties of Schwann cells and oligodendrocytes, transcriptional regulation of remyelination exhibits unique features and differs between the two cell types. This review gives an overview of the current state in the field.
Collapse
Affiliation(s)
- Elisabeth Sock
- Institut für Biochemie, Emil-Fischer-Zentrum, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Michael Wegner
- Institut für Biochemie, Emil-Fischer-Zentrum, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| |
Collapse
|
42
|
Elbaz B, Popko B. Molecular Control of Oligodendrocyte Development. Trends Neurosci 2019; 42:263-277. [PMID: 30770136 DOI: 10.1016/j.tins.2019.01.002] [Citation(s) in RCA: 196] [Impact Index Per Article: 32.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2018] [Revised: 01/07/2019] [Accepted: 01/15/2019] [Indexed: 12/27/2022]
Abstract
Myelin is a multilayer lipid membrane structure that wraps and insulates axons, allowing for the efficient propagation of action potentials. During developmental myelination of the central nervous system (CNS), oligodendrocyte progenitor cells (OPCs) proliferate and migrate to their final destination, where they terminally differentiate into mature oligodendrocytes and myelinate axons. Lineage progression and terminal differentiation of oligodendrocyte lineage cells are under tight transcriptional and post-transcriptional control. The characterization of several recently identified regulatory factors that govern these processes, which are the focus of this review, has greatly increased our understanding of oligodendrocyte development and function. These insights are critical to facilitate efforts to enhance OPC differentiation in neurological disorders that disrupt CNS myelin.
Collapse
Affiliation(s)
- Benayahu Elbaz
- The Center for Peripheral Neuropathy, The Department of Neurology, University of Chicago, Chicago, IL, USA
| | - Brian Popko
- The Center for Peripheral Neuropathy, The Department of Neurology, University of Chicago, Chicago, IL, USA.
| |
Collapse
|
43
|
Dolati S, Ahmadi M, Aghebti-Maleki L, Nikmaram A, Marofi F, Rikhtegar R, Ayromlou H, Yousefi M. Nanocurcumin is a potential novel therapy for multiple sclerosis by influencing inflammatory mediators. Pharmacol Rep 2018; 70:1158-1167. [DOI: 10.1016/j.pharep.2018.05.008] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2017] [Revised: 04/03/2018] [Accepted: 05/23/2018] [Indexed: 01/06/2023]
|
44
|
Klum S, Zaouter C, Alekseenko Z, Björklund ÅK, Hagey DW, Ericson J, Muhr J, Bergsland M. Sequentially acting SOX proteins orchestrate astrocyte- and oligodendrocyte-specific gene expression. EMBO Rep 2018; 19:embr.201846635. [PMID: 30166336 DOI: 10.15252/embr.201846635] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2018] [Revised: 07/24/2018] [Accepted: 08/07/2018] [Indexed: 12/30/2022] Open
Abstract
SOX transcription factors have important roles during astrocyte and oligodendrocyte development, but how glial genes are specified and activated in a sub-lineage-specific fashion remains unknown. Here, we define glial-specific gene expression in the developing spinal cord using single-cell RNA-sequencing. Moreover, by ChIP-seq analyses we show that these glial gene sets are extensively preselected already in multipotent neural precursor cells through prebinding by SOX3. In the subsequent lineage-restricted glial precursor cells, astrocyte genes become additionally targeted by SOX9 at DNA regions strongly enriched for Nfi binding motifs. Oligodendrocyte genes instead are prebound by SOX9 only, at sites which during oligodendrocyte maturation are targeted by SOX10. Interestingly, reporter gene assays and functional studies in the spinal cord reveal that SOX3 binding represses the synergistic activation of astrocyte genes by SOX9 and NFIA, whereas oligodendrocyte genes are activated in a combinatorial manner by SOX9 and SOX10. These genome-wide studies demonstrate how sequentially expressed SOX proteins act on lineage-specific regulatory DNA elements to coordinate glial gene expression both in a temporal and in a sub-lineage-specific fashion.
Collapse
Affiliation(s)
- Susanne Klum
- Ludwig Institute for Cancer Research, Karolinska Institutet, Stockholm, Sweden.,Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden
| | - Cécile Zaouter
- Ludwig Institute for Cancer Research, Karolinska Institutet, Stockholm, Sweden.,Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden
| | - Zhanna Alekseenko
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden
| | - Åsa K Björklund
- National Bioinformatics Infrastructure Sweden, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Daniel W Hagey
- Ludwig Institute for Cancer Research, Karolinska Institutet, Stockholm, Sweden.,Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden
| | - Johan Ericson
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden
| | - Jonas Muhr
- Ludwig Institute for Cancer Research, Karolinska Institutet, Stockholm, Sweden .,Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden
| | - Maria Bergsland
- Ludwig Institute for Cancer Research, Karolinska Institutet, Stockholm, Sweden .,Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
45
|
Fauveau M, Wilmet B, Deboux C, Benardais K, Bachelin C, Temporão AC, Kerninon C, Nait Oumesmar B. SOX17 transcription factor negatively regulates oligodendrocyte precursor cell differentiation. Glia 2018; 66:2221-2232. [PMID: 30152028 DOI: 10.1002/glia.23483] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Revised: 05/16/2018] [Accepted: 06/08/2018] [Indexed: 11/08/2022]
Abstract
Oligodendrocyte development is a critical process timely and spatially regulated to ensure proper myelination of the central nervous system. HMG-box transcription factors are key regulators of oligodendrocyte lineage progression. Among these factors, Sox17 was previously identified as a positive regulator of oligodendrocyte development. However, the role of Sox17 in oligodendroglial cell lineage progression and differentiation is still poorly understood. To define the functional role of Sox17, we generated new transgenic mouse models with inducible overexpression of Sox17, specifically in oligodendroglial cells. Here, we report that gain of Sox17 function has no effect on oligodendrocyte progenitor cells (OPCs) specification. During early postnatal development, Sox17 overexpression increases the pool of OPCs at the expense of differentiated oligodendrocytes. However, the oligodendroglial cell population, OPC proliferation and apoptosis remained unchanged in Sox17 transgenic mice. RNA sequencing, quantitative RT-PCR and immunohistochemical analysis showed that Sox17 represses the expression of the major myelin genes, resulting in a severe CNS hypomyelination. Overall, our data highlight an unexpected role for Sox17 as a negative regulator of OPC differentiation and myelination, suggesting stage specific functions for this factor during oligodendroglial cell lineage progression.
Collapse
Affiliation(s)
- Melissa Fauveau
- Inserm U1127, Institut du Cerveau et de la Moelle Epinière, ICM, Paris, France.,Sorbonne Université UMR-S1127, Paris, France.,CNRS, UMR 7225, Paris, France
| | - Baptiste Wilmet
- Inserm U1127, Institut du Cerveau et de la Moelle Epinière, ICM, Paris, France.,Sorbonne Université UMR-S1127, Paris, France.,CNRS, UMR 7225, Paris, France
| | - Cyrille Deboux
- Inserm U1127, Institut du Cerveau et de la Moelle Epinière, ICM, Paris, France.,Sorbonne Université UMR-S1127, Paris, France.,CNRS, UMR 7225, Paris, France
| | - Karelle Benardais
- Inserm U1127, Institut du Cerveau et de la Moelle Epinière, ICM, Paris, France.,Sorbonne Université UMR-S1127, Paris, France.,CNRS, UMR 7225, Paris, France
| | - Corinne Bachelin
- Inserm U1127, Institut du Cerveau et de la Moelle Epinière, ICM, Paris, France.,Sorbonne Université UMR-S1127, Paris, France.,CNRS, UMR 7225, Paris, France
| | - Ana C Temporão
- Inserm U1127, Institut du Cerveau et de la Moelle Epinière, ICM, Paris, France.,Sorbonne Université UMR-S1127, Paris, France.,CNRS, UMR 7225, Paris, France
| | - Christophe Kerninon
- Inserm U1127, Institut du Cerveau et de la Moelle Epinière, ICM, Paris, France.,Sorbonne Université UMR-S1127, Paris, France.,CNRS, UMR 7225, Paris, France
| | - Brahim Nait Oumesmar
- Inserm U1127, Institut du Cerveau et de la Moelle Epinière, ICM, Paris, France.,Sorbonne Université UMR-S1127, Paris, France.,CNRS, UMR 7225, Paris, France
| |
Collapse
|
46
|
Buitrago-Delgado E, Schock EN, Nordin K, LaBonne C. A transition from SoxB1 to SoxE transcription factors is essential for progression from pluripotent blastula cells to neural crest cells. Dev Biol 2018; 444:50-61. [PMID: 30144418 DOI: 10.1016/j.ydbio.2018.08.008] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2018] [Revised: 08/10/2018] [Accepted: 08/21/2018] [Indexed: 01/30/2023]
Abstract
The neural crest is a stem cell population unique to vertebrate embryos that gives rise to derivatives from multiple embryonic germ layers. The molecular underpinnings of potency that govern neural crest potential are highly conserved with that of pluripotent blastula stem cells, suggesting that neural crest cells may have evolved through retention of aspects of the pluripotency gene regulatory network (GRN). A striking difference in the regulatory factors utilized in pluripotent blastula cells and neural crest cells is the deployment of different sub-families of Sox transcription factors; SoxB1 factors play central roles in the pluripotency of naïve blastula and ES cells, whereas neural crest cells require SoxE function. Here we explore the shared and distinct activities of these factors to shed light on the role that this molecular hand-off of Sox factor activity plays in the genesis of neural crest and the lineages derived from it. Our findings provide evidence that SoxB1 and SoxE factors have both overlapping and distinct activities in regulating pluripotency and lineage restriction in the embryo. We hypothesize that SoxE factors may transiently replace SoxB1 factors to control pluripotency in neural crest cells, and then poise these cells to contribute to glial, chondrogenic and melanocyte lineages at stages when SoxB1 factors promote neuronal progenitor formation.
Collapse
Affiliation(s)
- Elsy Buitrago-Delgado
- Dept. of Molecular Biosciences, Northwestern University, Evanston, IL 60208, United States
| | - Elizabeth N Schock
- Dept. of Molecular Biosciences, Northwestern University, Evanston, IL 60208, United States
| | - Kara Nordin
- Dept. of Molecular Biosciences, Northwestern University, Evanston, IL 60208, United States
| | - Carole LaBonne
- Dept. of Molecular Biosciences, Northwestern University, Evanston, IL 60208, United States; Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Evanston, IL 60208, United States.
| |
Collapse
|
47
|
Santos AK, Vieira MS, Vasconcellos R, Goulart VAM, Kihara AH, Resende RR. Decoding cell signalling and regulation of oligodendrocyte differentiation. Semin Cell Dev Biol 2018; 95:54-73. [PMID: 29782926 DOI: 10.1016/j.semcdb.2018.05.020] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2018] [Revised: 05/15/2018] [Accepted: 05/17/2018] [Indexed: 12/20/2022]
Abstract
Oligodendrocytes are fundamental for the functioning of the nervous system; they participate in several cellular processes, including axonal myelination and metabolic maintenance for astrocytes and neurons. In the mammalian nervous system, they are produced through waves of proliferation and differentiation, which occur during embryogenesis. However, oligodendrocytes and their precursors continue to be generated during adulthood from specific niches of stem cells that were not recruited during development. Deficiencies in the formation and maturation of these cells can generate pathologies mainly related to myelination. Understanding the mechanisms involved in oligodendrocyte development, from the precursor to mature cell level, will allow inferring therapies and treatments for associated pathologies and disorders. Such mechanisms include cell signalling pathways that involve many growth factors, small metabolic molecules, non-coding RNAs, and transcription factors, as well as specific elements of the extracellular matrix, which act in a coordinated temporal and spatial manner according to a given stimulus. Deciphering those aspects will allow researchers to replicate them in vitro in a controlled environment and thus mimic oligodendrocyte maturation to understand the role of oligodendrocytes in myelination in pathologies and normal conditions. In this study, we review these aspects, based on the most recent in vivo and in vitro data on oligodendrocyte generation and differentiation.
Collapse
Affiliation(s)
- A K Santos
- Departamento de Bioquímica e Imunologia, Instituto de Ciência Biológicas, Universidade Federal de Minas Gerais, Av. Antônio Carlos, 6627, 31270-901 Belo Horizonte, MG, Brazil
| | - M S Vieira
- Departamento de Bioquímica e Imunologia, Instituto de Ciência Biológicas, Universidade Federal de Minas Gerais, Av. Antônio Carlos, 6627, 31270-901 Belo Horizonte, MG, Brazil; Instituto Nanocell, Rua Santo Antônio, 420, 35500-041 Divinópolis, MG, Brazil
| | - R Vasconcellos
- Departamento de Bioquímica e Imunologia, Instituto de Ciência Biológicas, Universidade Federal de Minas Gerais, Av. Antônio Carlos, 6627, 31270-901 Belo Horizonte, MG, Brazil; Instituto Nanocell, Rua Santo Antônio, 420, 35500-041 Divinópolis, MG, Brazil
| | - V A M Goulart
- Departamento de Bioquímica e Imunologia, Instituto de Ciência Biológicas, Universidade Federal de Minas Gerais, Av. Antônio Carlos, 6627, 31270-901 Belo Horizonte, MG, Brazil
| | - A H Kihara
- Centro de Matemática, Computação e Cognição, Universidade Federal do ABC, São Bernardo do Campo, SP, Brazil
| | - R R Resende
- Departamento de Bioquímica e Imunologia, Instituto de Ciência Biológicas, Universidade Federal de Minas Gerais, Av. Antônio Carlos, 6627, 31270-901 Belo Horizonte, MG, Brazil; Instituto Nanocell, Rua Santo Antônio, 420, 35500-041 Divinópolis, MG, Brazil.
| |
Collapse
|
48
|
Uncovering the Role of Sox2 in Oligodendroglia. J Neurosci 2018; 38:4460-4461. [PMID: 29743345 DOI: 10.1523/jneurosci.0556-18.2018] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Revised: 04/05/2018] [Accepted: 04/12/2018] [Indexed: 01/20/2023] Open
|
49
|
Cerrato V, Mercurio S, Leto K, Fucà E, Hoxha E, Bottes S, Pagin M, Milanese M, Ngan CY, Concina G, Ottolenghi S, Wei CL, Bonanno G, Pavesi G, Tempia F, Buffo A, Nicolis SK. Sox2 conditional mutation in mouse causes ataxic symptoms, cerebellar vermis hypoplasia, and postnatal defects of Bergmann glia. Glia 2018; 66:1929-1946. [PMID: 29732603 DOI: 10.1002/glia.23448] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2017] [Revised: 04/09/2018] [Accepted: 04/11/2018] [Indexed: 11/07/2022]
Abstract
Sox2 is a transcription factor active in the nervous system, within different cell types, ranging from radial glia neural stem cells to a few specific types of differentiated glia and neurons. Mutations in the human SOX2 transcription factor gene cause various central nervous system (CNS) abnormalities, involving hippocampus and eye defects, as well as ataxia. Conditional Sox2 mutation in mouse, with different Cre transgenes, previously recapitulated different essential features of the disease, such as hippocampus and eye defects. In the cerebellum, Sox2 is active from early embryogenesis in the neural progenitors of the cerebellar primordium; Sox2 expression is maintained, postnatally, within Bergmann glia (BG), a differentiated cell type essential for Purkinje neurons functionality and correct motor control. By performing Sox2 Cre-mediated ablation in the developing and postnatal mouse cerebellum, we reproduced ataxia features. Embryonic Sox2 deletion (with Wnt1Cre) leads to reduction of the cerebellar vermis, known to be commonly related to ataxia, preceded by deregulation of Otx2 and Gbx2, critical regulators of vermis development. Postnatally, BG is progressively disorganized, mislocalized, and reduced in mutants. Sox2 postnatal deletion, specifically induced in glia (with GLAST-CreERT2), reproduces the BG defect, and causes (milder) ataxic features. Our results define a role for Sox2 in cerebellar function and development, and identify a functional requirement for Sox2 within postnatal BG, of potential relevance for ataxia in mouse mutants, and in human patients.
Collapse
Affiliation(s)
- Valentina Cerrato
- Department of Neuroscience Rita Levi-Montalcini, University of Torino, Neuroscience Institute Cavalieri Ottolenghi (NICO), Regione Gonzole, 10, Orbassano, (Torino), 10043, Italy
| | - Sara Mercurio
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, piazza della Scienza 2, Milano, 20126, Italy
| | - Ketty Leto
- Department of Neuroscience Rita Levi-Montalcini, University of Torino, Neuroscience Institute Cavalieri Ottolenghi (NICO), Regione Gonzole, 10, Orbassano, (Torino), 10043, Italy
| | - Elisa Fucà
- Department of Neuroscience Rita Levi-Montalcini, University of Torino, Neuroscience Institute Cavalieri Ottolenghi (NICO), Regione Gonzole, 10, Orbassano, (Torino), 10043, Italy
| | - Eriola Hoxha
- Department of Neuroscience Rita Levi-Montalcini, University of Torino, Neuroscience Institute Cavalieri Ottolenghi (NICO), Regione Gonzole, 10, Orbassano, (Torino), 10043, Italy
| | - Sara Bottes
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, piazza della Scienza 2, Milano, 20126, Italy
| | - Miriam Pagin
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, piazza della Scienza 2, Milano, 20126, Italy
| | - Marco Milanese
- Department of Pharmacy, Pharmacology and Toxicology Unit and Center of Excellence for Biomedical Research, University of Genova, Viale Cembrano 4, Genoa, 16148, Italy
| | - Chew-Yee Ngan
- The Jackson Laboratory for Genomic Medicine, Farmington, Connecticut
| | - Giulia Concina
- Department of Neuroscience Rita Levi-Montalcini, University of Torino, Neuroscience Institute Cavalieri Ottolenghi (NICO), Regione Gonzole, 10, Orbassano, (Torino), 10043, Italy
| | - Sergio Ottolenghi
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, piazza della Scienza 2, Milano, 20126, Italy
| | - Chia-Lin Wei
- The Jackson Laboratory for Genomic Medicine, Farmington, Connecticut
| | - Giambattista Bonanno
- Department of Pharmacy, Pharmacology and Toxicology Unit and Center of Excellence for Biomedical Research, University of Genova, Viale Cembrano 4, Genoa, 16148, Italy
| | - Giulio Pavesi
- Department of Biosciences, University of Milano, 20100, Italy
| | - Filippo Tempia
- Department of Neuroscience Rita Levi-Montalcini, University of Torino, Neuroscience Institute Cavalieri Ottolenghi (NICO), Regione Gonzole, 10, Orbassano, (Torino), 10043, Italy
| | - Annalisa Buffo
- Department of Neuroscience Rita Levi-Montalcini, University of Torino, Neuroscience Institute Cavalieri Ottolenghi (NICO), Regione Gonzole, 10, Orbassano, (Torino), 10043, Italy
| | - Silvia K Nicolis
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, piazza della Scienza 2, Milano, 20126, Italy
| |
Collapse
|
50
|
Zhang S, Rasai A, Wang Y, Xu J, Bannerman P, Erol D, Tsegaye D, Wang A, Soulika A, Zhan X, Guo F. The Stem Cell Factor Sox2 Is a Positive Timer of Oligodendrocyte Development in the Postnatal Murine Spinal Cord. Mol Neurobiol 2018; 55:9001-9015. [PMID: 29623612 DOI: 10.1007/s12035-018-1035-7] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2017] [Accepted: 03/23/2018] [Indexed: 12/25/2022]
Abstract
Myelination in the central nervous system takes place predominantly during the postnatal development of humans and rodents by myelinating oligodendrocytes (OLs), which are differentiated from oligodendrocyte progenitor cells (OPCs). We recently reported that Sox2 is essential for developmental myelination in the murine brain and spinal cord. It is still controversial regarding the role of Sox2 in oligodendroglial lineage progression in the postnatal murine spinal cord. Analyses of a series of cell- and stage-specific Sox2 mutants reveal that Sox2 plays a biphasic role in regulating oligodendroglial lineage progression in the postnatal murine spinal cord. Sox2 controls the number of OPCs for subsequent differentiation through regulating their proliferation. In addition, Sox2 regulates the timing of OL differentiation and modulates the rate of oligodendrogenesis. Our experimental data prove that Sox2 is an intrinsic positive timer of oligodendroglial lineage progression and suggest that interventions affecting oligodendroglial Sox2 expression may be therapeutic for overcoming OPC differentiation arrest in dysmyelinating and demyelinating disorders.
Collapse
Affiliation(s)
- Sheng Zhang
- Institute for Pediatric Regenerative Medicine, Shriners Hospitals for Children/UC Davis School of Medicine, Sacramento, CA, 95817, USA.,Department of Neurology, School of Medicine, UC Davis, Davis, CA, 95817, USA
| | - Abeer Rasai
- Institute for Pediatric Regenerative Medicine, Shriners Hospitals for Children/UC Davis School of Medicine, Sacramento, CA, 95817, USA
| | - Yan Wang
- Institute for Pediatric Regenerative Medicine, Shriners Hospitals for Children/UC Davis School of Medicine, Sacramento, CA, 95817, USA.,Department of Neurology, School of Medicine, UC Davis, Davis, CA, 95817, USA
| | - Jie Xu
- Institute for Pediatric Regenerative Medicine, Shriners Hospitals for Children/UC Davis School of Medicine, Sacramento, CA, 95817, USA
| | - Peter Bannerman
- Institute for Pediatric Regenerative Medicine, Shriners Hospitals for Children/UC Davis School of Medicine, Sacramento, CA, 95817, USA.,Department of Cell Biology and Human Anatomy, School of Medicine, UC Davis, Davis, CA, 95817, USA
| | - Daffcar Erol
- Institute for Pediatric Regenerative Medicine, Shriners Hospitals for Children/UC Davis School of Medicine, Sacramento, CA, 95817, USA
| | - Danayit Tsegaye
- Institute for Pediatric Regenerative Medicine, Shriners Hospitals for Children/UC Davis School of Medicine, Sacramento, CA, 95817, USA
| | - Aijun Wang
- Institute for Pediatric Regenerative Medicine, Shriners Hospitals for Children/UC Davis School of Medicine, Sacramento, CA, 95817, USA.,Department of Surgery, School of Medicine, UC Davis, Davis, CA, 95817, USA
| | - Athena Soulika
- Institute for Pediatric Regenerative Medicine, Shriners Hospitals for Children/UC Davis School of Medicine, Sacramento, CA, 95817, USA.,Department of Dermatology, School of Medicine, UC Davis, Davis, CA, 95817, USA
| | - Xiangjiang Zhan
- Key Laboratory of Animal Ecology and Conservation Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Fuzheng Guo
- Institute for Pediatric Regenerative Medicine, Shriners Hospitals for Children/UC Davis School of Medicine, Sacramento, CA, 95817, USA. .,Department of Neurology, School of Medicine, UC Davis, Davis, CA, 95817, USA. .,Department of Neurology, UC Davis School of Medicine, c/o Shriners Hospitals for Children, Room 601A, 2425 Stockton Blvd, Sacramento, CA, 95817, USA.
| |
Collapse
|